Next Article in Journal
Effect of Ionization Degree of Poly(amidoamine) Dendrimer and 5-Fluorouracil on the Efficiency of Complex Formation—A Theoretical and Experimental Approach
Next Article in Special Issue
Anticoagulant Properties of Coated Fe-Pd Ferromagnetic Shape Memory Ribbons
Previous Article in Journal
Four-Dimensional Printing and Shape Memory Materials in Bone Tissue Engineering
Previous Article in Special Issue
Biological Cover Mitigates Disruption of the Dermal Structure in Mechanically Expanded Skin in a Porcine Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Application of Biomaterials in Spinal Cord Injury

Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(1), 816; https://doi.org/10.3390/ijms24010816
Submission received: 28 November 2022 / Revised: 19 December 2022 / Accepted: 30 December 2022 / Published: 3 January 2023

Abstract

:
The spinal cord and the brain form the central nervous system (CNS), which is the most important part of the body. However, spinal cord injury (SCI) caused by external forces is one of the most difficult types of neurological injury to treat, resulting in reduced or even absent motor, sensory and autonomic functions. It leads to the reduction or even disappearance of motor, sensory and self-organizing nerve functions. Currently, its incidence is increasing each year worldwide. Therefore, the development of treatments for SCI is urgently needed in the clinic. To date, surgery, drug therapy, stem cell transplantation, regenerative medicine, and rehabilitation therapy have been developed for the treatment of SCI. Among them, regenerative biomaterials that use tissue engineering and bioscaffolds to transport cells or drugs to the injured site are considered the most promising option. In this review, we briefly introduce SCI and its molecular mechanism and summarize the application of biomaterials in the repair and regeneration of tissue in various models of SCI. However, there is still limited evidence about the treatment of SCI with biomaterials in the clinic. Finally, this review will provide inspiration and direction for the future study and application of biomaterials in the treatment of SCI.

1. Introduction

The spinal cord is an important part of the human central nervous system and plays an extremely important role in the body’s reflex, conduction, motor and regulatory functions. Among the lesions of the spinal cord, spinal cord injury (SCI) is a central nervous system defect caused by traffic accidents, external violence and falls from height. With the continuous development of the global economy, the incidence of SCI shows an increasing trend. In the United States, more than 1 million people suffer from SCI, with more than 12,000 new cases each year [1]. The annual prevalence of SCI in China is 37 per million, with a mean age range of 34.7–54.4 years and a higher prevalence among males than females [2]. SCI not only brings endless pain and pressure to the patient itself but also imposes a serious burden on the patient’s family and society. SCI is difficult to treat and has gradually become a medical problem of global concern.
In recent years, with the continuous development of cell biology, regenerative medicine, and engineering materials science, tissue engineering technology has gradually become a new way to treat SCI. Cells, scaffolds, and growth factors are three key materials for tissue engineering. Cells and growth factors are usually implanted into the scaffolds and then play a therapeutic role in the injury site, while biomaterials are usually used as scaffolds, which can not only satisfy the attachment, delivery and growth of cells but can also mimic the natural extracellular matrix (ECM), which facilitates the binding of cells to tissues [3]. The basic approach is to construct artificial tissues in vitro with different bioscaffold materials, cells, neurotrophic factors, etc., so that the cells can adapt to the internal environment of the organism and the stimulation of biological factors and then be transplanted into the body to repair the damaged tissue [4]. Regarding the application of tissue engineering in the treatment of SCI, we summarize the properties, advantages, and biological activities of various biomaterials in this review. We hope this review provides insights for the future study of biomaterial scaffolds in the treatment of SCI.

2. Spinal Cord Injury

Spinal cord injury (SCI) is a serious complication of spinal fractures, including cervical, thoracic, conus medullaris and cauda equina injuries. It is characterized by high morbidity, disability, and mortality [5]. SCI generally manifests as limb sensory, motor, and autonomic dysfunction below the level of injury, which seriously hinders the patient’s daily life [6]. SCI patients with limb paralysis are prone to pulmonary infection, pressure ulcers, abnormal body temperature, etc., and the higher the injury site is, the more prone to complications, such as cardiovascular, respiratory and genitourinary complications, deep vein thrombosis, and chronic neuropathic pain, which may pose a serious threat to the patient’s life [7,8].
The pathogenesis of SCI is divided into primary injury and secondary injury. Primary injury is direct damage that generally has a physical impact on the spinal cord, mainly neuron and glial cell death, nerve axon and glial membrane destruction, blood vessel rupture with hemorrhage, etc. SCI is the main determinant of the severity of the initial extent of damage and the duration of spinal cord compression [9]. When the spinal cord is mechanically damaged, it can cause spinal cord compression, spinal cord contusion and concussion, axonal injury, etc. Primary SCI is a permanent irreversible dysfunction, and there is no effective clinical treatment yet [10]. Secondary injury usually occurs after the primary injury by a variety of factors, and the extent of damage sometimes exceeds the primary injury. Secondary injury is divided into the acute phase, subacute phase, and chronic phase. Acute clinical manifestations include increased cell permeability, apoptotic signal transduction, ischemia, vascular injury, edema, excitotoxicity, ion imbalance, inflammation, lipid peroxidation, and necrotic cell death [9]. Due to the destruction of the blood-spinal cord barrier, inflammatory cells, including macrophages, microglia, T cells, and neutrophils, infiltrate the injured site and trigger the release of many inflammatory cytokines, such as tumor necrosis factor α (TNF-α), interleukin (IL)-1α, IL-1β, and IL-6 [11], leading to organelle damage and oxidative cell death. Chondroitin sulfate proteoglycans (CSPG), as a component of the neuronal extracellular matrix network, are also up-regulated after SCI, which may be related to inflammatory response [12]. The subacute phase is followed by apoptosis, demyelination of surviving axons, Wallerian degeneration, axonal dieback, matrix remodeling, and the evolution of glial scarring around the injury site. The chronic phase will occur six months after SCI and is characterized by cyst formation, axonal retraction, and maturation of glial scars [13]. In addition, SCI can cause the proliferation of astrocytes and microglia in the nervous system to form glial scars, change the microenvironment of cell survival, increase proinflammatory cytokines, and imbalance the immune response [14] (Figure 1).
At present, the main treatments for SCI include surgery, drug-targeted therapy, stem cell therapy, biomaterials, tissue engineering scaffolds, and regenerative medicine [15] (Figure 2). Surgical treatment can solve the problems of spinal instability and fracture fragment displacement caused by primary injury. That is, surgery reduces the displaced vertebral body or removes the fracture fragments that protrude into the spinal canal to maintain the stability of the spine or to relieve the fracture to minimize further damage [16]. Dimar et al. used adult male rats to conduct decompression surgery in the early stage of SCI. The results showed that surgery can reduce SCI and is conducive to the recovery of neurological function [17]. In addition, Li et al. found that the paraspinal muscle approach combined with the posterior median approach effectively treats thoracolumbar fracture combined with SCI. This treatment has the advantages of a short operation time, minimal trauma, less bleeding and rapid recovery of patients after the operation [18]. Surgical treatment also has some defects. Patients have to bear postoperative physiological pain, and generated wounds due to surgery can easily damage the physiological environment of the body and cause infection. Early implementation of decompressive surgery has limited efficacy in complete SCI. In addition, many drugs, including hormone drugs, gangliosides, nerve growth factors, and traditional Chinese medicine (TCM), are also often used for the treatment of SCI. Among them, methylprednisolone (MP), a synthetic glucocorticoid, inhibits SCI-induced apoptosis by reducing the expression of the low-affinity nerve growth factor receptor p75 (p75NGFR) [19]. Studies have shown that gangliosides have a protective effect on damaged nerve cells and reduce further pathological changes after nerve damage via inhibiting apoptosis [10]. Nerve growth factor (NGF) can promote nerve cell differentiation and survival and axonal growth, which promotes the repair of damaged neurons, maintains the survival of mature neurons and promotes the regeneration of axons [20]. Hormonal drugs are prone to adverse reactions, and excessive doses cause other complications. The ingredients of TCS are complex and the mechanism of action is not clear. After SCI, a large number of axon cells and glial cells are lost and glial scarring prevents cell regeneration. Emerging evidence indicates that stem cell therapy has the characteristics of nerve regeneration, neuroprotection, and immune regulation, so stem cell therapy is also considered to be one of the most promising treatments in regenerative medicine [21]. To date, many stem cell types, including mesenchymal stem cells, olfactory ensheathing cells (OECs), Schwann cells (SCs), oligodendrocyte progenitor cells (OPCs), neural stem cells (NSCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs), have been intensively studied and tested in clinical trials [22]. Experimental studies have shown that exosomes derived from adipose-derived mesenchymal stem cells (ADMSCs) change the polarization direction of macrophages in rats with SCI and inhibit the formation of glial scarring [23]. The use of Schwann cell (SC) implantation into the injured spinal cord reduces cyst formation in the injured tissue, alleviates secondary damage to the tissue surrounding the initial injury site, and moderately improves limb movement [24]. Clinical studies have found that stem cells and drugs can be used to improve the SCI microenvironment to treat chronic SCI, and iPSCs introduced into the chronic SCI site can reduce cavitation and support the survival of transplanted cells [25].Although stem cell transplantation has a wide range of clinical applications, there are problems with stem cell therapy. Cell transplantation may induce lesions in other organs, block blood vessels, induce tumors, and cause rejection in the body, and the mechanism of action of stem cell transplantation is unclear. Thus, it is very important to find the best treatment for SCI. Recently, biomaterials have been used as carriers of cells or drugs due to the unique physicochemical properties, providing more treatment options for SCI.
Compared with other treatment methods, biomaterials are of great value in treating and repairing damaged tissues and assisting in the delivery and release of drugs. Drug delivery through biomaterials can reduce the problems of large invasion and blockage of direct drug administration. Although there are many biomaterials used in SCI in preclinical experiments, there are few generalizations about the characteristics of biomaterials and few review studies about the application of biomaterials in SCI. Therefore, this article reviews various types of biomaterials, focusing on various material properties, treatment modalities, etc. It provides a reference for biomaterials in the clinical treatment of SCI and provide directions for further scientific research.

3. The Application of Biomaterials in SCI

Biomaterials has been an emerging discipline in recent years, the study of which involves materials science, chemistry, medicine, pharmacology, etc. Biomaterials, also known as biomedical materials, are a class of natural or synthetic materials that can diagnose, repair, treat and induce regeneration of cells, tissues and organs and function in contact with body tissues or body fluids while causing fewer adverse reactions to the organism. Thus, biomaterials are widely used in clinical medicine, biotechnology and many other fields [26]. In the past, biomaterials were mainly used to replace damaged tissues due to diseases or trauma. However, in the last two decades, advances in materials manufacturing and characterization, the evolution of medical regeneration strategies and breakthrough successes in cellular and molecular biology and genetics have provided the substrate for the development of novel biomaterials with innovative applications. Emerging biomaterials aim not only to restore the structure and function of damaged tissues but also to regenerate them through active and targeted interactions [27]. Current biomaterials have been developed over a long period of time, as evidenced by the use of shells and gold for teeth and the use of twine for wound closure in the past, to the recent emergence of bioceramics, alkali metals, and polymeric materials. After several generations of development in the twenty-first century, biomaterials are now widely used in many fields, such as bone plates, artificial joints, vascular grafts, and nerve conduits [28]. The combination of biomaterials with cutting-edge technologies, such as nanotechnology, 3D printing technology, and neural stem cell technology, has brought biomaterials into the era of intelligent nanobiomaterials. On the other hand, the discipline of biomaterials has become the frontier of biomaterial research in drug delivery, tumor targeting therapy, tissue engineering, nanopreparation, bionic design, etc. [29,30,31]. Biomaterials have a wide range of applications and many varieties and are currently divided into natural biomaterials and synthetic biomaterials. Natural biomaterials mainly include chitosan, hyaluronic acid, fibrin, collagen, etc. [32]. The advantages of natural biomaterials are mainly their excellent biocompatibility, low immunogenicity, and nontoxic degradation [33]. In addition, synthetic materials have emerged and are widely used. In general, synthetic biomaterials, mainly polyethylene glycol (PEG), polylactic acid (PLA), polylactic acid-hydroxyacetic acid copolymer (PLGA), polyacrylamide (PAM), polyvinyl alcohol (PVA), and polymethyl methacrylate (PMMA), are attractive for their strong mechanical properties, customizable structure, and low immunogenicity. Their properties are often easier to tune than those of natural biomaterials. For example, the porosity, stiffness and degradation rates of synthetic biomaterials can be altered to match different types of tissues (Figure 3). The benchmark for the use of biomaterials is mainly their safety and performance. Therefore, biomaterials should have biocompatibility, biodegradability, and chemical stability to avoid rejection between materials and organisms.

3.1. Natural Materials

Natural materials are usually available from nature and are nontoxic, most of which include nucleic acids, polysaccharides, proteins, lipids and complex macromolecules. Biological activity can be imparted to materials through the use of natural polymers of nonmammalian and mammalian origin, which can be self-assembled or cross-linked to encapsulate natural tissue properties to form noncytotoxic hydrogels and scaffolds [34] (Table 1).

3.1.1. Hyaluronic Acid

Hyaluronic acid (HA), a widely studied and modified natural polymer for scaffolds, is a polysaccharide composed mainly of neurons and astrocytes, and astrocytes can synthesize high molecular weight HA [66,67]. HA is present in the extracellular matrix and is a major component of the extracellular matrix (ECM), playing an important role in the homeostasis of neuronal tissue by influencing cellular behaviors such as cell migration, value addition, and differentiation [68,69,70]. HA can be cross-linked between polymer chains and form highly tunable scaffolds through a variety of simple chemical modifications [71]. In addition, HA reduces glial scar formation by inhibiting the migration, chemotaxis, and proliferation of lymphocytes. It has been shown that HA affects the value-added of astrocytes, thus playing an important role in harmful glial scar formation, especially with high molecular weight HA [72,73]. After chemical modification and further processing and fabrication, 3D HA scaffolds are generated [74]. Covalently linking hydrogels to HA regulates cell growth and function in vitro, indicating that HA hydrogels modulate cell behavior. For example, astrocytes are affected by HA hydrogels; thus, astrocytes are a common target for HA hydrogel-based therapy [71]. In the treatment of SCI, HA hydrogels significantly reduced the level of glial fibrillary acidic protein (GFAP), indicating the level of active astrocytes, leading to the inhibition of harmful glial scar formation [36,75]. In addition, HA hydrogels improve the therapeutic effect of SCI by transporting drugs and stem cells to the injury area without affecting the cells. Emerging evidence indicates that neural stem cells (NSCs) generate new neurons and glia and secrete neuroprotective factors and growth factors to promote cell repair at the site of injury [76]. When using cell therapy, many cells are diluted or washed away, so keeping the cells at the injury site is the premise of treatment. However, HA hydrogel scaffolds can effectively retain cells in the injury site [77]. To improve the survival of transplanted neural stem cells and the integration of host tissues, adult brain-derived neural stem/progenitor cells (NSPCs) were implanted into hydrogel blends of HA and methyl cellulose (HAMC) modified with recombinant rat platelet-derived growth factor-A (rPDGF-A), which were shown to promote oligodendrocyte differentiation, improve graft survival, and reduce cavitation to improve behavioral capacity. In addition, the interaction between HA and NSCs contributes to the normal function of the adult CNS and the repair of damage [78]. Furthermore, HA hydrogels enhance the survival and expected differentiation of NSCs and improve motor function in SCI rats [35]. Therefore, HA is a useful scaffold that increases the stability and survival of implanted cells to promote SCI repair and improve motor ability.

3.1.2. Collagen

Collagen is one of the most abundant and widely distributed proteins in the human body. It is produced by fibroblasts and is the main component of the ECM, providing tensile strength for tissue growth and stimulating wound healing at the site of injury [79]. Because collagen resembles the ECM in the human body, immune rejection is lower when using biomaterials with composite collagen. In addition, the fibrous structure of collagen provides binding sites to support cell adhesion, migration, differentiation, and proliferation [80,81]. Therefore, collagen has gradually become one of the mainstream biomaterials for the treatment of SCI. In early studies, it was found that collagen acted as a carrier of neurotrophic factors to promote the repair of damaged areas [82]. For example, linear rat tail collagen (LRTC) was used as a carrier for recombinant collagen-bound neurotrophin-3 (CBD-NT3), which was found to restore some motor function and promote axonal regeneration after spinal cord transection damage. Currently, to deliver drugs, cells, proteins, and other substances to SCI injury areas, collagen can be designed into various types of scaffolds, such as sponges and hydrogels, and guide catheters [37]. Breen et al. injected a combination of neurotrophic factor (NT-3) and collagen hydrogel into a hemisected SCI model and found that neuronal and axonal growth was improved, while the inflammatory response and glial scar formation were inhibited [38]. Since collagen alone has poor mechanical strength and thermal stability, it is often used in combination with polymers. Wang et al. reported that collagen-binding vascular endothelial growth factor (CBD-VEGF) promoted axonal regeneration and cardiovascular generation in SCI injury sites [39]. Moreover, the N-calmodulin-modified linearly ordered collagen scaffold (LOCS) was shown to improve the mechanical adhesion of endogenous neural/progenitor stem cells (NSPC) to the scaffold, leading to neuronal regeneration and the improvement of motor function in SCI-transected rats [40]. In addition, the transplantation of LOCS and human placenta-derived mesenchymal stem cells (hPMSCs) improved neuronal regeneration and increased operational function in SCI-transected beagle dogs [41]. In addition, an increasing number of studies have shown that gelatin, a product of partial hydrolysis of collagen, also has therapeutic effects on SCI. It is a large molecule hydrocolloid that is usually combined with stem cells to form a scaffold to treat SCI [83]. Methacrylate-based gelatin (GelMA) hydrogels are very similar to natural extracellular matrixes (ECM) in some basic properties. One study showed that combining GelMA hydrogels with induced pluripotent stem cell (iPSC)-derived NSCs (iNSCs) significantly promotes the proliferation and facilitates functional recovery of neural stem cells [43]. In addition, GelMA can be fabricated into 3D materials, and loading BMSCs and NSCs into 3D GelMA hydrogels provides excellent mechanical properties for stem cell proliferation, migration, and differentiation [44]. Furthermore, GelMA and acrylated β-cyclodextrin can be formed into a supramolecular bioink (SM bioink), which then loads with an O-GlcNAc transferase (OGT) inhibitor and NSCs. The results showed that this scaffold promotes neuronal regeneration and axonal growth in an injury model [84]. Moreover, a sodium alginate/gelatin 3D scaffold encapsulated with neural stem cells and oligodendrocytes significantly improves hindlimb movement and nerve regeneration [45]. In summary, gelatin and collagen is a promising strategy for SCI repair mainly by combining it with stem cells and many neurotrophic factors while designing GelMA as a 3D or hydrogel scaffold material in terms of its role in the promotion of neural stem cell proliferation, migration, and differentiation.

3.1.3. Fibrin

Fibrin is a protein multimer that is mainly derived from plasma proteins and has good biocompatibility [45]. Injection of fibrin into the SCI can fill the lesion and provide a vehicle for implantation of stem cells, drugs and cytokines. Fibronectin can serve as a carrier for individual peripheral blood nuclei, and its implantation into a porcine SCI model promotes tissue repair in the immediate area of injury and restoration of conduction function in the posterior column of the spinal cord [46]. The fibrin hydrogel prepared by thrombin could smoothly implant nasopharyngeal carcinoma cells into the SCI site by providing cell attachment sites and survival signals and effectively alleviate the immune response at the SCI site [85]. To mimic the natural environment of spinal cord tissue, Yao et al. prepared a 3D hierarchically arranged fibrin hydrogel (AFG) that promotes targeted host cell invasion, vascular system reconstruction and axonal regeneration in SCI rat lesions [47]. In addition, promoting the differentiation of human endometrial stem cells to oligodendrocyte progenitors by upregulating miR-219 expression levels and using fibrin hydrogel as a scaffold to deliver human endometrial stem cells to the site of SCI significantly promoted the recovery of motor function in SCI rats [48]. The use of adipose mesenchymal stem cells combined with fibrin matrix reduces the cavity area, enhances tissue retention, inhibits the activation of astrocytes, and improves the microenvironment at the SCI site [86]. He et al. used fibrin hydrogels to deliver exosomes overexpressing VGF, the pro-SCI recovery regulator, and they found that the growth of oligodendrocytes in vivo and in vitro is improved [87]. Therefore, fibrin is usually designed as a hydrogel scaffold, which promotes the regeneration of neural tissue and the recovery of motor function by combining different factors.

3.1.4. Decellularized Scaffold

The decellularized matrix can be made into a porous natural biomaterial scaffold, obtained by removing nerve cells from spinal cord tissue and then freeze-drying them. Decellularized tissue retains the natural ECM components of the spinal cord after decellularization, which contains a large number of signaling molecules that regulate cell development, differentiation, and regeneration [88]. Decellularized tissue removes proteins associated with pathological factors such as the inflammatory response and scar formation and acts as an external microenvironment for implantation into the lesion, optimizing the imbalanced microenvironment after SCI, stimulating axonal regeneration, and promoting injury repair [89]. Cerqueira et al. injected cell-free injectable peripheral nerve matrix (iPN) into the lesions of spinal cord contusions, which successfully established immune tolerance and promoted the survival and axonal growth of SC in the transplanted bodies [49]. Cornelison et al. found that decellularized nerve grafts retain extracellular matrix components such as collagen and glycosaminoglycans, which reduce the ratio of M1:M2 macrophages in a rat cervical contusion model and facilitate axonal growth and nerve tissue repair [90]. It was demonstrated in in vitro experiments that in the structure of a rat spinal cord extracellular matrix scaffold, the decellular scaffold retains type IV collagen, fibronectin, laminin and other components in a three-dimensional meshwork structure with good pore space, which can promote the attachment and proliferation of bone marrow mesenchymal stem cells [91]. Liu et al. showed that decellularized spinal cord scaffolds can be used for polylactic acid–hydroxyacetic acid copolymer microspheres loaded with the pro-axonal growth drug bisperoxovanadium(pic) (bpV(pic)), which promote the viability of cultured neural stem cells and axonal growth by inhibiting phosphatase gene expression and activating the mTORC1/AKT pathway in vitro, as well as accelerate axonal regeneration and functional recovery in rats with SCI [50]. In addition, decellularized scaffolds can also be used to deliver adipose-derived stem cells (ADSCs), which effectively promotes histopathological repair and axonal regeneration, reduces reactive glial cell proliferation, and promotes functional recovery in SCI rats [51]. Therefore, decellularized scaffolds are often used to improve the internal microenvironment of SCI and reduce macrophage infiltration and cavity area.

3.1.5. Chitosan

Chitosan, a polysaccharide derived from chitin, is a positively charged natural biopolymer. Due to its unique properties, such as biocompatibility, biodegradability, adhesion, low toxicity, and the ability to form gels, it is widely used and is reported to have analgesic, antitumor, antibacterial, and hemostatic effects [92,93]. Moreover, the physicochemical properties of chitosan are modifiable, especially by altering and controlling its average acetylation degree (DA). Therefore, chitosan is considered a good biomaterial and has been widely investigated and used in various fields, including drug carriers [94], wound healing agents [95], lung surface active additives, and tissue engineering scaffolds [96]. In traumatic SCI, plasma membrane barrier function is degenerated, and neurons are lost or undergo necrosis [97]. At the same time, the passage of intracellular and extracellular ionic substances through this region leads to rapid conduction block within the nerve fibers. In addition, damage to the membrane may cause secondary damage and produce free radicals, which is accompanied by abnormal molecular signaling, inflammation, immune response, apoptosis, vascular changes, secondary cellular dysfunction, and finally, the production of potent endogenous toxins [98,99]. At the injury site of SCI, glial scar formation, neuronal and oligodendrocyte death, and the upregulation of axonal growth inhibitory factors were detected [100]. Therefore, to avoid the progressive destruction of cells, the barrier function of the damaged membrane needs to be re-established and resorted. Emerging evidence indicates that chitosan can be used to seal the damaged nerve cell membrane, thus serving as an effective neuroprotective agent after acute SCI and as an effective treatment for secondary damage [101]. In vitro and in vivo models of SCI have shown that chitosan is effective in regenerating neural tissue, restoring membrane integrity, and reducing the production of free radicals such as reactive oxygen species (ROS). In addition, autografts combined with biomaterials are a promising idea to allow for increased potential for axonal regeneration and functional recovery after SCI [102]. Chedly et al. reported that fragmented physical hydrogel suspensions (FPHS) containing chitosan and water as fragments had a defined degree of acetylation (DA), polymer concentration, and average fragment size. The implantation of FPHS reduced fibroglial scarring, promoted the reconstruction of the vascular system and spinal cord tissue, and modulated astrocyte responses and inflammatory responses at the site of SCI [52]. In addition, human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) and NT3-containing chitosan scaffolds attenuated the inflammatory response and microglial activation, promoted neuronal regeneration, facilitated neurological recovery, and improved motor function in SCI [53]. A chitosangelatin composite scaffold containing HA and acetyl heparan sulfate (HS) enhanced the adhesion and long-term expansion of NSCs and progenitor cells and the differentiation of NSCs and oligodendrocytes to neurons and glial cells [103]. Although great progress has been made in the use of chitosan-based scaffolds in the treatment of SCI, there is still a long way to go for their clinical application.

3.1.6. Alginate

Alginate is a hydrophilic long-chain polysaccharide that exists in the cell wall of algae, and its structure is flexible and gelatinous. Alginate exhibits a gel state when exposed to water, and its gel state can produce a series of 3D structures that facilitate drug transport and wound healing. Its unique chemical composition brings it closer to the physical properties of mammalian ECM than other natural organisms [104]. Therefore, alginate is widely used in the development of axon growth-promoting and targeted drug delivery. Although alginate can promote nerve regeneration under certain conditions, its mechanical strength is insufficient due to the fast degradation rate and a certain immune response. Thus, alginate is commonly used in combination with other natural polymers [105]. Hosseini et al. reported that the encapsulation of neural stem cells into alginate 3D scaffolds inhibited inflammation and reduced lesion size [54]. Covalently cross-linked lyophilized alginate enhanced neural regeneration in the spinal cord, and alginate served as a scaffold to promote regenerative axonal growth and the prolongation of astroglial processes [42]. In addition, alginate was designed as a 3D scaffold to help reconstruct ECM soft tissue at the damaged site by filling the injury-induced cavity. The implantation of a poly-L-ornithine-laminin-alginate (PLO-LAM-ALG) hydrogel scaffold promoted cell migration and slight axonal and nerve axon growth in the presence of cationic peptides [55]. Therefore, alginate is an ideal natural material as a 3D scaffold for the encapsulation of cells and factors in the treatment of SCI.

3.1.7. Agarose

In addition to to sodium alginate and chitosan, agarose is also a natural carbohydrate polymer and has excellent biocompatibility, thermally reversible gel behavior, excellent mechanical properties, and high bioactivity. Therefore, agarose has been widely used in tissue engineering and targeted drug delivery [106,107]. In early studies, different mechanical stiffnesses of agarose gels were found to affect the 3D neural protrusion elongation of sensory ganglia [108]. In subsequent studies, agarose hydrogels could guide 3D cell migration and nerve protrusion growth, and the transplantation of neurotrophic factors into multiluminal catheters promoted axonal regeneration after SCI [109,110]. In addition, templated agarose scaffolds supported linear axonal growth through their uniaxial channels and guided axonal regeneration after SCI [111,112]. Meanwhile, templated agarose could be made into linear arrays for the treatment of the post-damage spinal cord, and templated agarose multichannel scaffolds containing BDNF were reported to enhance nerve regeneration [56,113]. Composite electrodes made of agarose and carbon nanotube fibers (A-CNF) were effective in reducing the activity of astrocytes after the use of anti-inflammatory proteins [114]. In recent studies, the implantation of matrix gel into agarose scaffolds improved motility and promoted cell proliferation and axonal regeneration in SCI [57]. In addition, Yang et al. developed a unique agarose, gelatin, polypyrrole (Aga/Gel/PPy, AGP3) hydrogel, and in vitro results showed that AGP3 induced the differentiation of neural stem cells while inhibiting astrocyte formation and activating endogenous nerve regeneration in the spinal cord, resulting in significant restoration of motor function [58]. Therefore, templated agarose scaffolds serving as natural carbohydrate polymers are useful for the implantation of stem cells and the treatment of SCI.

3.1.8. Nanomaterials

The blood-brain barrier and the blood-spinal cord barrier interfere with the effective concentration of drugs entering the brain to some extent, thus requiring larger doses of drugs to achieve a certain therapeutic effect, which may lead to toxicity and side effects due to the increased concentration of drugs [115]. With unique structures, such as small size, large surface area, and large surface area ratio, nanomaterials can be used as drug carriers to deliver therapeutic drugs to targeted locations, reducing the effect in unwanted areas and improving bioavailability while reducing the occurrence of side effects. In addition, nanoparticles also modulate axonal regeneration and restore signal conduction in the injured spinal cord [116]. For the onset of inflammation secondary to SCI, MP effectively alleviated the inflammatory response and limited the secretion of cytokines at the injury site [117]. However, treatment with MP has significant limitations due to its significant side effects, including thromboembolism, blood pressure changes, ionic disturbances, sepsis, pneumonia, wound infections, and acute corticosteroid myopathy due to its low drug selectivity, high systemic doses, and toxicity [118]. The incorporation of MP into nanoparticles could reduce and avoid these side effects, improve bioavailability, increase the efficiency of drug transport, and improve motor function [59,60]. In a further study, circulating monocytes and neutrophils were internalized by polylactide deglycoside (PLGA) nanoparticles according to their physicochemical properties, which downregulate proinflammatory factors, upregulate anti-inflammatory and regenerative genes, inhibit fibrosis and glial scar formation, promote the regeneration of axons, and improve motor function [61]. Due to their unique characteristics, nanoparticles can be used to improve the mechanical properties of hydrogels, increase surface reactivity, and improve the degree of drug release and bioavailability. The addition of nanoparticles to the hydrogel allows for more effective access to the tissue through the capillaries and more efficient delivery of therapeutic agents to the injury site [119,120]. Nerve tissue and cells have the unique property of generating and transmitting electrical stimuli, which affects not only neuronal firing but also cell proliferation, differentiation, and migration [4]. Therefore, electrical stimulation is a nonchemical method that can be applied to nerve regeneration. One of the most important aspects for the use of electrical stimulation to repair spinal cord injuries is the need to find a biocompatible electroactive biomaterial. Numerous studies have shown that the carbon-based nanomaterial graphene has excellent physicochemical and mechanical properties and good electrical conductivity, which allows it to use neural electrical signals in spinal cord tissue to stimulate axonal regeneration and promote the differentiation of stem cells. Graphene is a 2D material consisting of a single layer of carbon atoms arranged in a hexagonal honeycomb lattice, which has excellent properties such as high carrier mobility, quantum Hall effect at room temperature, high optical transparency, excellent mechanical strength, and excellent thermal conductivity [121,122,123]. Thus, graphene has a wide range of applications in various fields, such as sensors [124], supercapacitors [125], composite materials, cancer targeting, photothermal therapy, drug delivery, and tissue engineering. In addition, due to its large surface area and its ability to be surface functionalized, graphene can also be used as a nanocarrier to transport drugs, proteins, etc. Among them, graphene oxide (GO), a member of the graphene family, is widely used for the treatment of SCI because of its large loading, hydrophilic functional groups and excellent biocompatibility. In a recent study, GO was used in combination with polyethylene glycol (PEG) and chitosan (CS) to form novel nanocomposites, which promoted cell growth, reduced inflammatory responses, and improved motor function [62]. Three-dimensional graphene, also known as graphene foam, has been well used in tissue repair. Studies have shown that 3D graphene with better biocompatibility can stimulate and accelerate the growth and differentiation of nerve cells in axonal repair and inhibit neuroinflammation [126,127]. In addition to classical graphene nanoparticles, nanoparticles such as metal nanoparticles and polymer nanoparticles also exist. Metallic nanoparticles are currently showing the potential to design novel delivery systems, which can be divided into pure metal nanoparticles and metal oxide nanoparticles. They act on SCI by changing their shape and size, and are then modified by various types of chemical functional groups. Subsequently, modified metal nanoparticles can bind various drugs, antibodies, nutritional factors, etc. [128,129]. Zonisamide (1,2-benzisoxazole-3-methanesulfonamide) has been reported as an antiepileptic drug, but some studies have found that it can exert a certain therapeutic effect on neurological dysfunction. In Fang’s study, the use of zonisamide-loaded metal nanoparticles showed promise in modifying neurons and axons to promote recovery from SCI [130]. In addition, metal nanoparticles can also promote the immunogenicity of protein immunity. In one study, gold nanoparticles were used as adjuvants to enhance the activity of a 15 nm GNP-coupled human NgR-Fc (hNgR-Fc) protein vaccine, thereby promoting damage repair [131]. Another study showed that laser exposure of gold nanorods can promote the growth of reaching elements [132]. Therefore, in Mina’s study, the authors combined chondroitinase ABCI (cABCI) with different concentrations of gold nanorods. The results of the study exhibited better stability of the enzyme upon binding, thus reducing chondroitin sulfate proteoglycans (CSPG) and promote neuronal regeneration [133]. Moreover, polymeric nanoparticles have also been shown to be therapeutically effective in spinal cord injuries. Ling et al. investigated whether that combining poly(α-lipoic acid)-methylprednisolone (PαLA-MP) prodrug nanoparticles (NPs) and minocycline (MC) could produce better anti-inflammatory effects [134]. In addition, combining rapamycin with mesoporous polydopamine nanoparticles has good ROS clearing ability and exhibits reduced injury cavity, enhanced motility and promotes nerve regeneration in animal models [135]. Taken together, nanomaterials play a key role in carrying and transporting drugs or cells to the targeted tissue of SCI.

3.1.9. Self-Assembled Peptides

Self-assembled peptides (SAPs) are monomers composed of short or repetitive amino acid sequences that, when assembled, can form nanostructures that are attractive in the field of regeneration. By modifying their physicochemical properties and amino acid composition, SAPs perform a variety of biological functions and are more reactive than other conventional nonbiological materials. When composed of nano- or micro-meter structures, SAPs exhibit various abilities, such as tissue regeneration engineering, drug delivery, stability and targeting of drug release, and reduction of toxicity and side effects. The advantages of SAPs include biocompatibility, ease of synthesis, and ability for effective target recognition [136]. However, due to their poor metabolic stability and rapid clearance, SAPs are not always suitable as drugs [137]. In SCI, self-assembling peptide nanofiber scaffolds (SAPNS) can bridge the injured spinal cord, trigger axonal regeneration, inhibit the inflammatory response, and ultimately promote the recovery of motor function [138]. RADA(16)-IKVAV self-assembled peptide hydrogels were used as functional peptide-based scaffolds to load neural stem cells, which increased neural axon growth and reduced glial astrocyte formation [139]. In addition, self-assembling peptide gel (SPG-178) promoted the expression of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophic factor-4 (NT-4), and promyosin receptor kinases (TrkA and TrkB) in nerve synapses and inhibited neuroinflammation and glial scar formation [140]. Zhao et al. proposed that the combined injection of neural precursor cells and SAPs into damaged neural tissue could enhance neural repair and regeneration [63]. Wang et al. prepared a new FGLmx peptide hydrogel scaffold using pure RADA16 and found that it promoted the proliferation and migration of neural stem cells, thereby facilitating the repair of SCI [64]. In addition, the transplantation of microvessels into RADA-16 l reduced inflammation and glial scar formation and increased the density of axonal growth to the injury/graft site, which demonstrates the potential of vascularized scaffolds in the repair of SCI [65]. Therefore, although many studies have demonstrated the therapeutic role of SAPs, more research is required to increase their metabolic stability and reduce their clearance rate.

3.2. Synthetic Materials

Synthetic materials are widely used in tissue engineering and regenerative medicine for the treatment of SCI because they mimic the physicochemical and mechanical properties of cellular tissues. In addition, synthetic materials have low toxicity to the CNS and are reported to inhibit the inflammatory response and reduce glial scarring. Furthermore, the degradation mode of synthetic materials is simple hydrolysis, which facilitates their applications in tissue engineering [141,142]. However, the hydrolysis process has certain drawbacks in that it produces carbon dioxide, which lowers the local pH value and thus leads to cell and tissue necrosis, so more care should be taken when using these materials [143]. Among these synthetic materials, aliphatic polyesters are biodegradable materials, mainly including polylactic acid (PLA), polyglycolic acid (PGA), polycaprolactone (PCL) and their copolymers. They are well known for their high mechanical strength, flexibility, easy processing, and nontoxic degradability [144,145]. In addition, the blending of different types of polymers can form a unique scaffold material with its original properties and exhibit good therapeutic effects in various models, which has been a research direction of biomaterials (Table 2).

3.2.1. Polyethylene Glycol

Polyethylene glycol (PEG) is a nonionic water-soluble polymer that is a hydrolysis product of ethylene oxide and exhibits different physical properties depending on its molecular weight, which is widely used in food and drug applications as a solvent, surfactant, solubilizer, etc. [171]. In addition, PEG can reduce the adverse reaction of drugs by prolonging the duration of action and reducing the immunogenicity of drugs [172]. Thus, PEG is considered to be a safe and nontoxic polymer. When PEG is used in combination with hydrogels, the different molecular weights of PEG affect the mechanical properties of the hydrogels [171,173]. Currently, PEG is usually combined with nanoparticles to form PEGylated nanoparticles, which increase the circulation time, facilitate crossing the blood-brain barrier, and reduce the adverse effects of drugs [174,175]. A composite scaffold consisting of cell growth factor 2 (FGF2), epidermal growth factor (EGF), glial-derived neurotrophic factor (GDNF), PEG, and PCL significantly improved motor function and increased axonal regeneration [146]. In addition, PEG cross-linked with hydrogels has been widely used to improve the regeneration of axons after SCI due to its hydrophilicity, high water content, porosity [176], rejection of nonspecific protein adsorption and cell adhesion [177,178]. In addition, PEG hydrogels in a 3D environment can be used as drug delivery carriers to modulate inflammatory responses, promote axonal regeneration, facilitate molecular and cellular uptake and diffusion, and promote the differentiation of stem cells [179]. Loss of cell membrane integrity leads to cell death and the spread of damage to parts beyond the injury. In SCI, PEG inhibits free radical generation and resists the level of lipid peroxidation, indicating that PEG acts as a fusion agent to reseal the disrupted plasma membrane and protect mitochondria against injury [147]. Therefore, the key application of PEG serves as a fusion agent to repair the cell membrane at the injury site, leading to a reduction in oxidative stress, axonal regeneration, and the restoration of motor function [148,180]. However, it has been found that PEG is only effective in repairing cell membranes at higher concentrations, while 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG) could repair the dysfunction of SCI more effectively at lower concentrations [149]. Therefore, PEG is an ideal biomaterial that inhibits the disruption of the cell membrane to promote the regeneration of neurons in SCI.

3.2.2. Polylactic Acid

Polylactic acid (PLA) is made from renewable resources and is the most commonly used and promising renewable aliphatic polyester for the synthesis of biodegradable polymers [181]. PLA is now widely used in biomedical applications, such as tissue engineering scaffolds, drug delivery systems, implants, and coverings. In addition, unique properties, such as nontoxicity, bioresorbability, environmental friendliness, and thermoplasticity, make PLA and its lysis products biocompatible with SCs and spinal cord tissue [182,183]. Therefore, PLA can be designed into various types of scaffolds for the regeneration of tissue cells after SCI, including micron or nanofibers, hydrogels, microporous sponges, etc. [184,185]. However, PLA also has certain disadvantages, including poor toughness, slow degradation, and hydrophobicity [186,187], so it is usually used in combination with other polymers or nonpolymers. For example, PLA/PPy scaffolds formed by polylactic acid (PLA) and polypyrrole (PPy) composite could aid the transplantation of bone marrow stromal cells (BMSCs) in SCI, leading to the regeneration of neurons, the restoration of conduction, the inhibition of glial scar formation, and the regeneration of axons, which provides a better microenvironment for the treatment of SCI [150]. In addition, drug-loaded fiber mats made of core-shell nanofibers equipped with docosahexaenoic acid (DHA) in the core and PLA as the shell were prepared by electrostatic spinning, which has sufficient mechanical properties to promote the growth of neuronal synapses [151]. In another study, aligned PLA microfiber grafts without any cells, neurotrophic factors, or drugs promoted the regeneration of CNS tissues [152]. There, PLA has a wide range of application aspects and can be designed into different scaffold types to treat SCI.

3.2.3. Poly(Lactic-Co-Glycolic Acid)

Poly(lactic-co-glycolic acid) (PLGA) is formed by the polymerization of lactic acid and ethanol, which are degradable functional organic polymers. PLGA has different molecular weights and copolymer ratios, indicating that the ratio of the two monomers affects their physicochemical properties [188]. Studies have shown that PLGA has the following properties: slow controlled release, low toxicity, biodegradability, and biocompatibility [189]. Thus, PLGA can be used as a carrier for hydrophobic and hydrophilic substances [190,191]. To date, PLGA hydrogels, catheter scaffolds, and nanoparticles are the three most widely used drug delivery systems [192]. PLGA is usually used in combination with nanoparticles, and PLGA nanoparticles can increase the retention time of macromolecules in vivo and regulate the release rate and distribution of macromolecules [193]. Gwak et al. reported that PLGA nanospheres modified with 3β-[N-(N′,N′-dimethylaminoethane) carbamoyl] cholesterol PLGA/DC-Chol nanospheres improved the efficiency of gene transfection, promoted vascular and tissue regeneration, and improved the motility of SCI rats [153]. Moreover, PLGA/PLLA has been used as a composite scaffold to treat SCI in several ways. For example, the implantation of human oral mucosal stem cells (hOMSCs) into the PLGA/PLLA scaffold could promote endogenous repair by secreting neuroprotective, immunomodulatory, and axonal extension-related factors, thereby restoring motility and reducing glial scar formation [154]. In addition, PLGA/PLLA scaffolds implanted with human dental pulp stem cells (DPSCs) are highly vascularized and have great potential for tract vascular production and nerve regeneration in SCI rats [155]. It has been reported that the diameter and porosity of scaffolds have a significant impact on axonal regeneration of the damaged spinal cord, with smaller diameter scaffolds showing a better effect for SCI repair [194,195]. Therefore, designing PLAG multichannel catheters with hierarchical pore structures can enhance gene expression in vivo and promote value-added cell spreading, which can improve the repair of damaged spinal cords [196]. In addition, longitudinal porous scaffolds prepared by mixing HA hydrogels and PLGA have a better therapeutic effect on SCI. For example, the combinational use of anti-Nogo receptor antibodies (antiNgR) and PLGA microspheres containing BDNF and VEGF could inhibit inflammation and promote revascularization [156]. Furthermore, PLGA scaffolds can promote the differentiation of stem cells in the tissue of SCI [197], and the implantation of NSCs into PLGA scaffolds can effectively promote the recovery of neurological functions in vivo [198,199]. In conclusion, the development of PLGA scaffolds and their composite scaffolds with other polymers has become popular and widely used in the treatment of SCI, and more research on gene modification and optimization of the physical properties of the scaffold are required in the future.

3.2.4. Polycaprolactone

Polycaprolactone (PCL) is a highly elastic fatty acid polyester that is biocompatible and biodegradable, and it is commonly used in tissue engineering because of its slow degradation rate and low toxicity [200,201]. In a recent study, 3D-aligned PCL microfibrous scaffolds of different diameters were prepared, and their effects on the differentiation of stem/progenitor cells were investigated. The results showed that the differentiation of stem cells, astrocytes, and oligodendrocytes was increased by PCL scaffolds, suggesting that PCL is useful for the treatment of SCI [202]. The implantation of differentiated cells from hEnSCs cocultured with human Schwann cells (hSCs) onto PCL/gelatin scaffolds limited secondary damage, promoted axonal regeneration, and restored sensory and motor function after SCI [157]. To allow PCL to function as a nerve-guided scaffold with maximum open volume, the cell adhesion of PCL is improved by changing the porosity of PCL, resulting in the fabrication of porous PCL microtubular scaffolds, which have been demonstrated to promote axonal growth and reduce scar tissue in SCI rats [159]. In addition, Gelain et al. found that scaffolds containing PCL nanofibers and bioactive self-assembling peptides induced neural regeneration and angiogenesis and restored the motility of SCI rats [203]. Furthermore, a trimethylene carbonate and ε-caprolactone (TC) block copolymer catheter scaffold containing longitudinally microgrooved poly-p-dioxanone microfilaments (PDOs) was designed, and the results showed that the TC/PDO catheter promoted the migration of different cell types, such as SCs, while promoting the growth of sensory axons and attenuating the response of astrocytes to SCI injury [158]. Therefore, PCL scaffolds promote cell differentiation and improve the motor function of SCI.

3.2.5. Polyvinyl Alcohol (PVA)

Polyvinyl alcohol (PVA) is a nontoxic, biocompatible, and biodegradable hydrophilic polymer. In the treatment of SCI, PVA was found to significantly inhibit inflammatory responses and alleviate secondary damage. In an early study, the implantation of PVA hydrogel into the spinal cord prevented the migration of inflammatory cells and reduced the formation of scar tissue and adhesions [204]. In a subsequent study, PVA and PVP containing p38 and JNK inhibitors reduced the inflammatory response and decreased the expression of proteins associated with cell death [160]. In addition, Chen et al. prepared a composite molybdenum sulfide/graphene oxide/PVA (MoS 2/GO/PVA) hydrogel with good biocompatibility, electrical conductivity, and moderate mechanical properties. They found that this hydrogel could promote the differentiation of neurons, inhibit the inflammatory response, and gradually restore motor function [161]. In conclusion, research on PVA has mainly focused on its anti-inflammatory effect in SCI, and more research on the biological activity of PVA is needed.

3.2.6. Poly-Beta-Hydroxybutyrate

Poly-beta-hydroxybutyrate (PHB) is a biopolyester with a large molecular weight that exists in the cytoplasm of many bacteria and is a degradable biomaterial [205]. The transplantation of SCs into PHB catheters could better fill the cavity and significantly increase SC cell attachment, proliferation, and axon regeneration [206]. In recent years, Zhao et al. found that the blending of polyhydroxybutyrate-cohydroxyvalerate (PHBV) with PLA and collagen (Col) could improve the performance of the polymer. The prepared nanofiber scaffold significantly promotes the differentiation of astrocytes but inhibits their overactivation. It can also reduce CSPG and glial fibrillary acidic protein (GFAP) and promotes the recovery of motor function [162]. Another study showed that the implantation of PHBV/PLA/Col membrane and VSC4.1 motor neurons into rats with SCI can inhibit inflammasomes and reduce glial scar formation by reducing the infiltration of CD86-positive macrophages in the lesion and promoting axon growth [163]. In addition, NLRP3 inflammasome expression can be suppressed using the ketone metabolite β-hydroxybutyrate (βOHB), resulting in less neuroinflammation and better motor performance and electrophysiological recovery [164]. Doncel-Pérez et al. used poly(3-hydroxybutyrate-co-3-hydroxycaproic acid) fibrous scaffolds coated with laminin or polylysine/laminin to culture rat neural progenitor cells. Then, synchrotron radiation infrared microspectroscopy observation results showed that the neural progenitor cells had good adhesion and proliferation behavior [207]. Agrawal et al. developed a new type of biomaterial mixed melanin with PHB. This scaffold is compatible with nerve tissue in terms of physicochemical and electrical properties, and has rich surface nanotopography, semiconducting properties, and pore viscoelastic properties similar to brain tissue. Therefore, it can improve cell adhesion and growth of sensory and motor neurons in mice [208]. Therefore, the research on PHB can start from its mechanism of action and suitable composite materials.

3.2.7. Polysialic Acid

Polysialic acid (PSA), a class of linear homogeneous α-2,8 carbohydrates linked to sialic acid, is most prominent during nervous system development and can be combined with neural adhesion molecules in the vertebrate nervous system through typical n-linked glycosidic bonds [209,210]. PSA can induce the migration of SCs, which has a great effect on improving repair after SCI. Pearse et al. used the lentiviral vector-transduced mouse polysialyltransferase gene ST8SIA4 (LV-PST) and recombinant bacterial enzyme engineering (PSTNm) to upregulate the expression of PSA in nerve cells, proving that PSA can effectively regulate the immune response and improve the migration of SCs and neural repair after SCI [211]. A PSA-based minocycline nanodrug delivery system (PSM) exhibits significant anti-inflammatory and neuroprotective effects in vitro and in vivo, and PSM can recruit endogenous neural stem cells to the injury site in SCI rats, promoting neuronal regeneration and long axon extension throughout the glial scar to improve motor function [212]. Studies have shown that PCL/PSA hybrid nanofiber scaffolds encapsulated with MP implanted into the lesion area can effectively inhibit acute tissue inflammation and apoptosis and promote axon regeneration, thereby promoting nerve repair and improving functional prognosis [165]. In addition, the use of PSA ring mimetic peptide (PR-21) inhibits the hyperplasia of reactive glia and improves motor sensory function [213]. In Mehanna’s study, the combination of PSA and human natural killer cell-1 (HNK-1) glycan shows the poor effect in the early stage of injury but can promote axonal myelination and the recovery of function after 6 weeks [166]. Recently, a collagen laminin (C/L) scaffold was used to load the PSA analog compound 5-nonoxytryptamine (5-NOT), which promotes the survival of cortical neurons and neurite length and improves motor function through the ERK-MAPK pathway [167]. In conclusion, finding a suitable PSA mimic is an important direction for future research on SCI.

3.2.8. Poly(2-Hydroxyethyl Methacrylate)

Most of the current tissue-engineered scaffolds for the treatment of SCI are chosen from biodegradable natural or synthetic polymers because nondegradable or slowly degradable polymers require a postprocessing process, which makes treatments more difficult. Currently, nondegradable polymers such as acrylic acid have been gradually used in the treatment of SCI, of which poly(2-hydroxyethyl methacrylate) (pHEMA) is a flexible hydrophilic porous material with biocompatible and stable mechanical properties that can incorporate collagen and fibrin as a bioadhesive matrix [214]. Tsai et al. used a new synthetic hydrogel composite of PHEMA and methyl methacrylate (MMA) as a guiding channel and found that the PHEMA-MMA hydrogel guiding channel promoted axon regeneration and reduced glial scar formation in spinal cord transection injury [168,170]. In addition, PHEMA hydrogel scaffolds containing BDNF increased the number of axons and blood vessels and promoted recovery of motor function in SCI, accompanied by a mild inflammatory response [215]. A PHEMA hydrogel implanted carrying charged functional groups was reported to improve axonal regeneration at injury sites [216]. PHEMA lacks functional groups that support cell adhesion. After the immobilization of laminin-derived Ac-CGGGASIKVAVS-OH (SIKVAV) peptide and fibronectin subunit (Fn) on PHEMA hydrogel scaffolds, cell adhesion and cell proliferation and differentiation were improved [217]. Kubinova et al. optimized the porosity and mechanical properties of PHEMA hydrogels and found that a 68% porosity of PHEMA hydrogels had the best effect on tissue response and axonal growth in SCI rats [169]. In addition, the design of homogeneous and bilayer PHEMA hydrogel scaffolds has mechanical properties and biocompatibility, which are more suitable for spinal cord tissues [218]. Valdes-Sanchez et al. reported that caprolactone 2-(methacryloyloxy) ethyl ester (CLMA) made into a 3D scaffold after esterification from PHEMA and PCL could improve the survival of mature neurons after binding to acutely transplanted epithelial stem/progenitor cells (epSPCs) [219]. The use of SIKVAV-HEMA hydrogel scaffolds with oriented voids showed better infiltration in connective tissue and blood vessels but slower infiltration in axons, so further research is needed to support the type of scaffold in the microenvironment of the injury site [220]. Therefore, the modification or combinational use of pHEMA with other materials provides therapeutic options for SCI.

3.3. Titanium Alloys

Today, titanium and titanium-based alloys are also widely used biomaterials because of their good biocompatibility, low density, corrosion resistance, and good mechanical efficiency. At present, it is mainly used to replace orthopedic implant materials [221]. The supplement of Zr, V, and Mo elements to titanium can improve the mechanical properties of titanium, which manufactures bone plates and cardiovascular stents [222]. Research has shown that mesenchymal stem cells differentiate into osteoblasts and promote bone union and remodeling when four new alloys are implanted into rabbit models. Titanium-based biomaterials with tailored porosity can affect cell adhesion, differentiation, and growth [223]. Therefore, alloy materials can be considered for the treatment of SCI. When titanium-based alloy is used as a biomaterial for SCI repair, it needs to consider that the alloy may react with the liquid environment in the body to produce toxicity and trigger inflammation [224]. Therefore, how to make composite scaffolds from natural or synthetic materials and titanium-based alloys is a promising research direction in the future.

3.4. 3D Scaffold

Three-dimensional bioprinting technology allows the size and shape of the scaffold to be adjusted as needed. Many biomaterials are used to design materials as 3D scaffolds for repairing SCI. In a study, a novel biocompatible bioink consisting of functional chitosan, hyaluronic acid derivatives and matrix gels was found to rapidly gel and spontaneously covalently cross-link to maintain the viability of neural progenitor cells and effectively support axonal regeneration in a rat model of SCI [225]. Three-dimensional-printed scaffolds can transport homogeneous bone marrow MSCs and SCs in a specific spatial arrangement, which promote the formation of intercellular junctions and directed cell differentiation [226]. Cryoextrusion 3D printing technology can maintain the biological activity of the delivered cytokines. The collagen/chitosan scaffold prepared by low-temperature extrusion 3D printing technology successfully fuses brain-derived neurotrophic factors, which fills the injury gap, promotes nerve fiber regeneration, accelerates the establishment of synaptic connections, and promotes myelin regeneration at the injury site in rats with SCI [227]. Three-dimensional-printed collagen/silk fibroin scaffolds which mimic the structure of corticospinal tracts can guide the targeted repair of damaged neural tissue in rats [228]. Joung et al. prepared physiologically active neuronal networks by precisely placing clusters of spinal cord neuronal progenitor cells and oligodendrocyte progenitor cells within 3D printed biocompatible scaffolds during assembly and controlling the cell cluster positions using a distribution-by-dot printing method [229]. Koffler et al. used microscale continuous projection printing to create complex neural network structures and printed 3D bionic hydrogel scaffolds loaded with neural progenitor cells that supported axonal regeneration in SCI lesions in mice, induced neuronal synapses upstream and downstream of the implant to connect to neural progenitor cells within the scaffold, and significantly improved functional outcomes. Moreover, the scaffold is scalable to human spinal cord size and lesion geometry [230]. In summary, the 3D scaffold can fill the damaged part well by adjusting its shape and size.

4. Clinical Application of Biomaterial Scaffolds

Although many biomaterial scaffolds have been widely used in the study of SCI, few have been translated from preclinical experiments to clinical studies. Previously, Amr et al. investigated the repair effect of chitosan–laminin scaffold-wrapped MSCs on patients with chronic paraplegia. The results showed that motor function is recovered despite a gap in the spinal cord, the patients regain the ability of some muscle groups, and the thoracolumbar spine is improved. Moreover, the recovery process is not affected by the postoperative seroma formed by the disintegration of chitosan [231]. Xiao et al. studied the safety and feasibility of the collagen scaffold NeuroRegen after implantation into the human body. During the follow-up one year later, they found that there was no adverse reaction related to stent implantation and no more serious consequences such as deterioration of the nervous system. In addition, they observed partial recovery of the patient’s autonomic nervous function [232]. In a recent study, after the implantation of MSCs into NeuroRegen scaffolds, the subsequent follow-up found that the sensory function of the patient’s bowel and bladder recovered and the sensory level expanded below the injury site. In addition, some patients could control the activities of the toes, suggesting the recovery of some muscle tissue [233]. Chen et al. implanted NeuroRegen stents with autologous bone marrow mononuclear cells (BMMCs) into the body. In addition to stress pulmonary ulcer, pulmonary infection and transient high fever in the short term after the operation, no significant cystic cavitation or malignant proliferation was observed in the follow-up 3 years later. In addition, the patients exhibited expansion of the sensory level and recovery of autonomic nervous function [234]. In addition to the collagen scaffold NeuroRegen, a spinal cord scaffold composed of covalently conjugated poly(lactic acid-glycolic acid) and poly(l-lysine) InVivo Therapeutics Corp and CS collagen scaffolds have also been clinically studied [235,236]. These clinical studies have confirmed the clinical feasibility of biomaterials, but most of the current clinical trials remain in phase I studies, and the effectiveness of biomaterials in the treatment of SCI still faces great challenges. It is believed that more scaffold materials will be converted into clinical applications in the future.

5. Summary and Future Outlook

At present, SCI has become one of the most difficult central neurological disorders to treat, and patients usually show a decrease or loss of motor and sensory function below the injury site. However, the repair of this damage may be affected by many factors, such as apoptosis of neuronal cells, deposition of glial scarring, and inflammatory cascade response. This article mainly discusses various biomaterial scaffolds for SCI treatment, among which the relatively popular scaffolds are hydrogel and 3D scaffolds. Various materials are designed so that these scaffolds can simulate the internal environment of spinal cord tissue and can meet the repair of the injured part by adjusting the size and shape of the scaffold. Compared with traditional drug intervention and surgical treatment, the use of biomaterial scaffolds can reduce some complex side effects of drugs and obstacles to functional recovery after surgery. The physical and chemical properties of these biomaterials and the structure of the scaffold play an important role in the treatment of SCI. Compared with other treatments, we should pay more attention to the issues of biomaterials, such as biocompatibility, degradability, mechanical strength, and toxicity to peripheral tissues.
Although natural biomaterials mimic ECM and show good biocompatibility, due to their inherent characteristics, there are also some unavoidable shortcomings, such as insufficient scaffold strength, mismatch between cell degradation rate and regeneration rate, and scaffold collapse after the material swells with water. Among them, fibrin has poor mechanical strength and is easy to degrade, which is not conducive to long-term culture in vitro, and the arrangement of fibers lacks directionality, which reduces the induction effect on directional growth of axons. HA is easily soluble in water and absorbs and degrades too quickly. Alginate is prone to the immune response. Synthetic materials increase the hardness of the scaffold but lack inherent biological functions and must undergo significant postprocessing to trigger the desired response in vivo, and their degradation products are prone to local inflammatory responses that destroy the microenvironment, resulting in the decrease in cell survival. In addition, synthetic materials have weak affinity to cells, and are commonly used to prepare composite scaffolds with natural materials to induce nerve axon regeneration. For example, PCL has insufficient mechanical strength; PLA has a slow degradation rate and poor hydrophobicity; PSA has the disadvantage of being difficult to purify; PHB has poor toughness and a narrow processing time window. It is worth noting that regardless of what kind of biomaterial is implanted into the spinal cord, it cannot cause secondary damage to the body.
In conclusion, although these scaffold materials have made some progress in the treatment of SCI, there are still many problems to be solved. When using different scaffolds to treat SCI, the detailed mechanism of action still needs to be further studied, and how to combine the drugs, NTs, and stem cells loaded on the stents to achieve the best therapeutic effect still needs to be deeply understood. In addition, when using a scaffold for treatment, there is uncertainty about the optimal timing of implantation, location, and duration of treatment while the stent is in the body. In future research, the main research directions include the construction of a stable microenvironment, the study of therapeutic mechanisms, how to improve the shortcomings of various biomaterials, and the development and application of new composite biomaterials in order to find materials with the best therapeutic effect. Most importantly, future research should pay more attention to the safety and effectiveness evaluation of biomaterial scaffolds in clinical trials, so as to promote the application of SCI in clinical treatment.

Author Contributions

A.-G.W. and X.-G.Z. conceived the idea; C.F. and L.D. wrote the original manuscript; X.-G.Z., J.-M.W. and D.-L.Q. revised the manuscript; A.-G.W. and L.Y. created the figures; Y.-Y.Y. summarized the tables. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (Grant No. 81903829), the Department of Science and Technology of Sichuan Province, China (Grant Nos. 2020YJ0494, 2019JDJQ0033, and 22ZDYF3784), the Macao Science and Technology Development Fund of Macao SAR (Project Nos. SKL-QRCM(MUST)-2020-2022 and MUST-SKL-2021-005), Southwest Medical University (Grant Nos. 2022YFS0620-B4, 2021ZKZD015, 2021ZKZD018, and 2021ZKMS046), Luzhou Science and Technology Project, China (2022-SYF-73), and the joint project of Luzhou Municipal People’s Government and Southwest Medical University, China (Grant No. 2020LZXNYDJ37).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Hachem, L.D.; Ahuja, C.S.; Fehlings, M.G. Assessment and management of acute spinal cord injury: From point of injury to rehabilitation. J. Spinal Cord Med. 2017, 40, 665–675. [Google Scholar] [CrossRef] [PubMed]
  2. Chen, X.; Chen, D.; Chen, C.; Wang, K.; Tang, L.; Yuzhe, L.I.; Aimin, W.U. The epidemiology and disease burden of traumatic spinal cord injury in China: A systematic review. Chin. J. Evid. Based Med. 2018, 18, 143–150. [Google Scholar]
  3. Ikada, Y. Challenges in tissue engineering. J. R. Soc. Interface 2006, 3, 589–601. [Google Scholar] [CrossRef] [PubMed]
  4. Thompson, B.C.; Murray, E.; Wallace, G.G. Graphite Oxide to Graphene. Biomaterials to Bionics. Adv. Mater. 2015, 27, 7563–7582. [Google Scholar] [CrossRef] [PubMed]
  5. Yang Junsong, H.D. Research status and future of stem cell therapy for spinal cord injury. Chin. J. Trauma 2021, 37, 6–10. [Google Scholar] [CrossRef]
  6. Berg, M.V.D.; Castellote, J.; Mahillo-Fernandez, I.; de Pedro-Cuesta, J. Incidence of Spinal Cord Injury Worldwide: A Systematic Review. Neuroepidemiology 2010, 34, 184–192, discussion 192. [Google Scholar] [CrossRef] [PubMed]
  7. Hagen, E.M. Acute complications of spinal cord injuries. World J. Orthop. 2015, 6, 17–23. [Google Scholar] [CrossRef]
  8. Zou, X.; Jing, C.; Yingmei, Q.; Ying, L. Care of a patient with old spinal cord injury causing paraplegia combined with diabetic pressure sores. Jilin Med. 2011, 32, 6963–6964. [Google Scholar]
  9. Anjum, A.; Yazid, M.; Daud, M.F.; Idris, J.; Lokanathan, Y. Spinal Cord Injury: Pathophysiology, Multimolecular Interactions, and Underlying Recovery Mechanisms. Int. J. Mol. Sci. 2020, 21, 7533. [Google Scholar] [CrossRef]
  10. Jian, Q.I.; Zhang, J.J.; Meng, Q.X.; Zhao, T.B. Pathology and Treatment of Spinal Cord Injury. Prog. Mod. Biomed. 2017, 17, 4179–4183. [Google Scholar]
  11. Ahuja, C.S.; Satoshi, N.; Lindsay, T.; Jefferson, W.; Brian, K.; James, H.; David, C.; Fehlings, M.G. Traumatic Spinal Cord Injury—Repair and Regeneration. Neurosurgery 2017, 80, S9–S22. [Google Scholar] [CrossRef]
  12. Weiner, H.L. A shift from adaptive to innate immunity: A potential mechanism of disease progression in multiple sclerosis. J. Neurol. 2008, 255 (Suppl. S1), 3–11. [Google Scholar] [CrossRef] [PubMed]
  13. Dimitrijevic, M.R.; Danner, S.M.; Mayr, W. Neurocontrol of Movement in Humans With Spinal Cord Injury. Artif. Organs 2015, 39, 823–833. [Google Scholar] [CrossRef] [PubMed]
  14. Shen, H.; Fan, C.; You, Z.; Xiao, Z.; Zhao, Y.; Dai, J. Advances in Biomaterial-Based Spinal Cord Injury Repair. Adv. Funct. Mater. 2022, 32, 2110628. [Google Scholar] [CrossRef]
  15. 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]
  16. Pei, D. Research status of spinal cord injury at home and abroad. Everyone Healthy 2017, 12, 440–444. [Google Scholar]
  17. Dimar, J.R.; Glassman, S.D.; Raque, G.H.; Zhang, Y.P.; Shields, C.B. The Influence of Spinal Canal Narrowing and Timing of Decompression on Neurologic Recovery After Spinal Cord Contusion in a Rat Model. Spine 1999, 24, 1623. [Google Scholar] [CrossRef]
  18. Jing-Feng, L.I.; Zhen-Hua, X.U.; Chen, L.B.; Wang, J.P.; Zhang, S.W.; Orthopedics, D.O. Paraspinal approach combined with posterior midline approach in treatment of thoracolumbar fractures with nerve injury. Chin. J. Bone Jt. Inj. 2015, 30, 263–265. [Google Scholar]
  19. Mallei, A.; Aden, S.A.; Bachis, A.; Brandoli, C.; Ongini, E.; Mocchetti, I. The nitrosteroid NCX 1015, a prednisolone derivative, improves recovery of function in rats after spinal cord injury. Brain Res. 2005, 1062, 16–25. [Google Scholar] [CrossRef]
  20. Jinchuan, S.; Qingmin, F. Advances in research on neurotrophic factors and spinal cord injury. J. Binzhou Med. Coll. 2013, 36, 136–138. [Google Scholar]
  21. Marina, G.; Vladislav, V.; Harrell, C.; Crissy, F.; Nemanja, J.; Nebojsa, A.; Miodrag, S. Stem Cells Therapy for Spinal Cord Injury. Int. J. Mol. Ences 2018, 19, 1039. [Google Scholar]
  22. Haoxian, C.; Lijun, Y.; Yonggang, A. Effects of Exosomes Derived from Adipose-Derived Mesenchymal Stem cells on Macrophage Polarization and Glial Scar Formation in Rats with Spinal Cord Injury. Front. Mol. Neurosci. 2022, 43, 333–338. [Google Scholar] [CrossRef]
  23. Yamazaki, K.; Kawabori, M.; Seki, T.; Houkin, K. Clinical Trials of Stem Cell Treatment for Spinal Cord Injury. Int. J. Mol. Sci. 2020, 21, 3994. [Google Scholar] [CrossRef] [PubMed]
  24. Wiliams, R.R.; Bunge, M.B. Schwann cell transplantation: A repair strategy for spinal cord injury? Prog Brain Res. 2012, 201, 295–312. [Google Scholar] [CrossRef]
  25. 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]
  26. Marin, E.; Boschetto, F.; Pezzotti, G. Biomaterials and biocompatibility: An historical overview. J. Biomed. Mater. Res. Part A 2020, 108, 1617–1633. [Google Scholar] [CrossRef]
  27. Masaeli, R.; Zandsalimi, K.; Tayebi, L. Biomaterials Evaluation: Conceptual Refinements and Practical Reforms. Ther. Innov. Regul. Sci. 2019, 53, 120–127. [Google Scholar] [CrossRef]
  28. Ratner, B.D.; Zhang, G. A History of Biomaterials. In Biomaterials Science; Academic Press: Cambridge, MA, USA, 2020; pp. 21–34. [Google Scholar]
  29. Yuan, T.; Zhang, L.; Li, K.; Fan, H.; Fan, Y.; Liang, J.; Zhang, X. Collagen hydrogel as an immunomodulatory scaffold in cartilage tissue engineering. J. Biomed. Mater. Res. Part B Appl. Biomater. 2014, 102, 337–344. [Google Scholar] [CrossRef]
  30. Masand, S.N.; Chen, J.; Perron, I.J.; Hammerling, B.C.; Loers, G.; Schachner, M.; Shreiber, D.I. The effect of glycomimetic functionalized collagen on peripheral nerve repair. Biomaterials 2012, 33, 8353–8362. [Google Scholar] [CrossRef] [Green Version]
  31. 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]
  32. Kim, B.-S.; Baez, C.E.; Atala, A. Biomaterials for tissue engineering. World J. Urol. 2000, 18, 2–9. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, M.; Zhai, P.; Chen, X.; Schreyer, D.J.; Sun, X.; Cui, F. Bioengineered Scaffolds for Spinal Cord Repair. Tissue Eng. Part B Rev. 2011, 17, 177–194. [Google Scholar] [CrossRef] [PubMed]
  34. Joyce, K.; Fabra, G.T.; Bozkurt, Y.; Pandit, A. Bioactive potential of natural biomaterials: Identification, retention and assessment of biological properties. Signal Transduct. Target. Ther. 2021, 6, 122. [Google Scholar] [CrossRef] [PubMed]
  35. 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] [PubMed]
  36. 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]
  37. Fan, J.; Xiao, Z.; Zhang, H.; Chen, B.; Tang, G.; Hou, X.; Ding, W.; Wang, B.; Zhang, P.; Dai, J.; et al. Linear Ordered Collagen Scaffolds Loaded with Collagen-Binding Neurotrophin-3 Promote Axonal Regeneration and Partial Functional Recovery after Complete Spinal Cord Transection. J. Neurotrauma 2010, 27, 1671–1683. [Google Scholar] [CrossRef]
  38. 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]
  39. Wang, L.; Shi, Q.; Dai, J.; Gu, Y.; Feng, Y.; Chen, L. Increased vascularization promotes functional recovery in the transected spinal cord rats by implanted vascular endothelial growth factor-targeting collagen scaffold. J. Orthop. Res. 2018, 36, 1024–1034. [Google Scholar] [CrossRef] [Green Version]
  40. 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]
  41. Han, S.; Xiao, Z.; Li, X.; Zhao, H.; Wang, B.; Qiu, Z.; Mei, X.; Xu, B.; Fan, C.; Chen, B.; et al. Human placenta-derived mesenchymal stem cells loaded on linear ordered collagen scaffold improves functional recovery after completely transected spinal cord injury in canine. Sci. China Life Sci. 2018, 61, 2–13. [Google Scholar] [CrossRef]
  42. 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] [PubMed]
  43. 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] [PubMed]
  44. Zhou, P.; Xu, P.; Guan, J.; Zhang, C.; Chang, J.; Yang, F.; Xiao, H.; Sun, H.; Zhang, Z.; Wang, M.; et al. Promoting 3D neuronal differentiation in hydrogel for spinal cord regeneration. Colloids Surfaces B Biointerfaces 2020, 194, 111214. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, S.; Yang, H.; Chen, D.; Xie, Y.; Tai, C.; Wang, L.; Wang, P.; Bin Wang, B. Three-dimensional bioprinting sodium alginate/gelatin scaffold combined with neural stem cells and oligodendrocytes markedly promoting nerve regeneration after spinal cord injury. Regen. Biomater. 2022, 9, rbac038. [Google Scholar] [CrossRef] [PubMed]
  46. Shulman, I.; Ogurcov, S.; Kostennikov, A.; Rogozin, A.; Garanina, E.; Masgutova, G.; Sergeev, M.; Rizvanov, A.; Mukhamedshina, Y. Application of Autologous Peripheral Blood Mononuclear Cells into the Area of Spinal Cord Injury in a Subacute Period: A Feasibility Study in Pigs. Biology 2021, 10, 87. [Google Scholar] [CrossRef] [PubMed]
  47. 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]
  48. Monfared, M.J.; Nasirinezhad, F.; Ebrahimi-Barough, S.; Hasanzade, G.; Saberi, H.; Tavangar, S.M.; Asadpour, S.; Aryan, L.; Barabadi, Z.; Ai, J. Transplantation of miR-219 overexpressed human endometrial stem cells encapsulated in fibrin hydrogel in spinal cord injury. J. Cell. Physiol. 2019, 234, 18887–18896. [Google Scholar] [CrossRef]
  49. Cerqueira, S.R.; Lee, Y.-S.; Cornelison, R.C.; Mertz, M.W.; Wachs, R.A.; Schmidt, C.E.; Bunge, M.B. Decellularized peripheral nerve supports Schwann cell transplants and axon growth following spinal cord injury. Biomaterials 2018, 177, 176–185. [Google Scholar] [CrossRef]
  50. Liu, J.; Li, K.; Huang, K.; Yang, C.; Huang, Z.; Zhao, X.; Song, S.; Pang, T.; Zhou, J.; Wang, Y.; et al. Acellularized spinal cord scaffolds incorporating bpV(pic)/PLGA microspheres promote axonal regeneration and functional recovery after spinal cord injury. RSC Adv. 2020, 10, 18677–18686. [Google Scholar] [CrossRef]
  51. Yin, H.; Jiang, T.; Deng, X.; Yu, M.; Xing, H.; Ren, X. A cellular spinal cord scaffold seeded with rat adipose-derived stem cells facilitates functional recovery via enhancing axon regeneration in spinal cord injured rats. Mol. Med. Rep. 2018, 17, 2998–3004. [Google Scholar] [CrossRef]
  52. 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] [PubMed] [Green Version]
  53. Sun, G.D.; Shao, J.L.; Deng, D.J.; Zhou, Z.G.; Zhou, X.B.; Lin, Y.X.; Li, Z.Z. A chitosan scaffold infused with neurotrophin-3 and human umbilical cord mesenchymal stem cells suppresses inflammation and promotes functional recovery after spinal cord injury in mice. Int. J. Clin. Exp. Med. 2017, 10, 11672–11679. [Google Scholar]
  54. Hosseini, S.M.; Sharafkhah, A.; Koohi-Hosseinabadi, O.; Semsar-Kazerooni, M. Transplantation of Neural Stem Cells Cultured in Alginate Scaffold for Spinal Cord Injury in Rats. Asian Spine J. 2016, 10, 611–618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Schackel, T.; Kumar, P.; Günther, M.; Liu, S.; Brunner, M.; Sandner, B.; Puttagunta, R.; Müller, R.; Weidner, N.; Blesch, A.; et al. Peptides and Astroglia Improve the Regenerative Capacity of Alginate Gels in the Injured Spinal Cord. Tissue Eng. Part A 2019, 25, 522–537. [Google Scholar] [CrossRef]
  56. Gao, M.; Lu, P.; Lynam, D.; Bednark, B.; Campana, W.M.; Sakamoto, J.; Tuszynski, M. BDNF gene delivery within and beyond templated agarose multi-channel guidance scaffolds enhances peripheral nerve regeneration. J. Neural Eng. 2016, 13, 066011. [Google Scholar] [CrossRef]
  57. 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]
  58. 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]
  59. Kim, Y.-T.; Caldwell, J.-M.; Bellamkonda, R.V. Nanoparticle-mediated local delivery of methylprednisolone after spinal cord injury. Biomaterials 2009, 30, 2582–2590. [Google Scholar] [CrossRef]
  60. Cerqueira, S.R.; Oliveira, J.M.; Silva, N.A.; Leite-Almeida, H.; Ribeiro-Samy, S.; Almeida, A.; Mano, J.F.; Sousa, N.; Salgado, A.J.; Reis, R.L. Microglia Response and In Vivo Therapeutic Potential of Methylprednisolone-Loaded Dendrimer Nanoparticles in Spinal Cord Injury. Small 2013, 9, 738–749. [Google Scholar] [CrossRef]
  61. Park, J.; Zhang, Y.; Saito, E.; Gurczynski, S.J.; Moore, B.B.; Cummings, B.J.; Anderson, A.J.; Shea, L.D. Intravascular innate immune cells reprogrammed via intravenous nanoparticles to promote functional recovery after spinal cord injury. Proc. Natl. Acad. Sci. USA 2019, 116, 14947–14954. [Google Scholar] [CrossRef] [Green Version]
  62. Yari-Ilkhchi, A.; Ebrahimi-Kalan, A.; Farhoudi, M.; Mahkam, M. Design of graphenic nanocomposites containing chitosan and polyethylene glycol for spinal cord injury improvement. RSC Adv. 2021, 11, 19992–20002. [Google Scholar] [CrossRef] [PubMed]
  63. Zhao, X.; Yao, G.S.; Liu, Y.; Wang, J.; Satkunendrarajah, K.; Fehlings, M. The role of neural precursor cells and self assembling peptides in nerve regeneration. J. Otolaryngol. Head Neck Surg. 2013, 42, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wang, J.; Zheng, J.; Zheng, Q.; Wu, Y.; Wu, B.; Huang, S.; Fang, W.; Guo, X. FGL-functionalized self-assembling nanofiber hydrogel as a scaffold for spinal cord-derived neural stem cells. Mater. Sci. Eng. C Mater. Biol. Appl. 2015, 46, 140–147. [Google Scholar] [CrossRef] [PubMed]
  65. Tran, K.A.; Partyka, P.P.; Jin, Y.; Bouyer, J.; Fischer, I.; Galie, P.A. Vascularization of self-assembled peptide scaffolds for spinal cord injury repair. Acta Biomater. 2020, 104, 76–84. [Google Scholar] [CrossRef] [PubMed]
  66. Song, I.; Dityatev, A. Crosstalk between glia, extracellular matrix and neurons. Brain Res. Bull. 2018, 136, 101–108. [Google Scholar] [CrossRef]
  67. A Back, S.; Tuohy, T.M.F.; Chen, H.; Wallingford, N.; Craig, A.; Struve, J.; Luo, N.L.; Banine, F.; Liu, Y.; Chang, A.; et al. Hyaluronan accumulates in demyelinated lesions and inhibits oligodendrocyte progenitor maturation. Nat. Med. 2005, 11, 966–972. [Google Scholar] [CrossRef]
  68. Gupta, R.C.; Lall, R.; Srivastava, A.; Sinha, A. Hyaluronic Acid: Molecular Mechanisms and Therapeutic Trajectory. Front. Vet. Sci. 2019, 6, 192. [Google Scholar] [CrossRef] [Green Version]
  69. Moshayedi, P.; Carmichael, S.T. Hyaluronan, neural stem cells and tissue reconstruction after acute ischemic stroke. Biomatter 2013, 3, e23863. [Google Scholar] [CrossRef]
  70. Frischknecht, R.; Chang, K.-J.; Rasband, M.N.; Seidenbecher, C.I. Neural ECM molecules in axonal and synaptic homeostatic plasticity. Prog. Brain Res. 2014, 214, 81–100. [Google Scholar] [CrossRef]
  71. Collins, M.N.; Birkinshaw, C. Hyaluronic acid based scaffolds for tissue engineering—A review. Carbohydr. Polym. 2013, 92, 1262–1279. [Google Scholar] [CrossRef]
  72. Meng, F.; Modo, M.; Badylak, S.F. Biologic scaffold for CNS repair. Regen. Med. 2014, 9, 367–383. [Google Scholar] [CrossRef] [PubMed]
  73. Sensharma, P.; Madhumathi, G.; Jayant, R.D.; Jaiswal, A.K. Biomaterials and cells for neural tissue engineering: Current choices. Mater. Sci. Eng. C Mater. Biol. Appl. 2017, 77, 1302–1315. [Google Scholar] [CrossRef] [PubMed]
  74. Lee, J.U.; Kim, D.; Jang, C.H.; Kim, G.H. Highly elastic 3D-printed gelatin/HA/placental-extract scaffolds for bone tissue engineering. Theranostics 2022, 12, 4051–4066. [Google Scholar] [CrossRef] [PubMed]
  75. Hou, S.; Xu, Q.; Tian, W.; Cui, F.; Cai, Q.; Ma, J.; Lee, I.-S. The repair of brain lesion by implantation of hyaluronic acid hydrogels modified with laminin. J. Neurosci. Methods 2005, 148, 60–70. [Google Scholar] [CrossRef]
  76. Sabelström, H.; Stenudd, M.; Frisén, J. Neural stem cells in the adult spinal cord. Exp. Neurol. 2014, 260, 44–49. [Google Scholar] [CrossRef]
  77. 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]
  78. 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] [Green Version]
  79. Sorushanova, A.; Delgado, L.M.; Wu, Z.; Shologu, N.; Kshirsagar, A.; Raghunath, R.; Mullen, A.M.; Bayon, Y.; Pandit, A.; Raghunath, M.; et al. The Collagen Suprafamily: From Biosynthesis to Advanced Biomaterial Development. Adv. Mater. 2019, 31, e1801651. [Google Scholar] [CrossRef]
  80. Li, X.; Dai, J. Bridging the gap with functional collagen scaffolds: Tuning endogenous neural stem cells for severe spinal cord injury repair. Biomater. Sci. 2018, 6, 265–271. [Google Scholar] [CrossRef]
  81. Gu, L.; Shan, T.; Ma, Y.-X.; Tay, F.R.; Niu, L. Novel Biomedical Applications of Crosslinked Collagen. Trends Biotechnol. 2019, 37, 464–491. [Google Scholar] [CrossRef]
  82. Houweling, D.; Lankhorst, A.; Gispen, W.; Bär, P.; Joosten, E. Collagen Containing Neurotrophin-3 (NT-3) Attracts Regrowing Injured Corticospinal Axons in the Adult Rat Spinal Cord and Promotes Partial Functional Recovery. Exp. Neurol. 1998, 153, 49–59. [Google Scholar] [CrossRef] [PubMed]
  83. Taheri, A.; Kenari, A.A.; Gildberg, A.; Behnam, S. Extraction and Physicochemical Characterization of Greater Lizardfish (Saurida tumbil) Skin and Bone Gelatin. J. Food Sci. 2009, 74, E160–E165. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, X.; Song, S.; Chen, Z.; Gao, C.; Li, Y.; Luo, Y.; Huang, J.; Zhang, Z. Release of O-GlcNAc transferase inhibitor promotes neuronal differentiation of neural stem cells in 3D bioprinted supramolecular hydrogel scaffold for spinal cord injury repair. Acta Biomater. 2022, 151, 148–162. [Google Scholar] [CrossRef]
  85. Sudhadevi, T.; Vijayakumar, H.S.; Hariharan, E.V.; Sandhyamani, S.; Krishnan, L.K. Optimizing fibrin hydrogel toward effective neural progenitor cell delivery in spinal cord injury. Biomed. Mater. 2021, 17, 014102. [Google Scholar] [CrossRef]
  86. Mukhamedshina, Y.O.; Akhmetzyanova, E.; Kostennikov, A.; Zakirova, E.Y.; Galieva, L.R.; Garanina, E.E.; Rogozin, A.A.; Kiassov, A.P.; Rizvanov, A. Adipose-Derived Mesenchymal Stem Cell Application Combined With Fibrin Matrix Promotes Structural and Functional Recovery Following Spinal Cord Injury in Rats. Front. Pharmacol. 2018, 9, 343. [Google Scholar] [CrossRef] [Green Version]
  87. He, X.; Yang, L.; Dong, K.; Zhang, F.; Liu, Y.; Ma, B.; Chen, Y.; Hai, J.; Zhu, R.; Cheng, L. Biocompatible exosome-modified fibrin gel accelerates the recovery of spinal cord injury by VGF-mediated oligodendrogenesis. J. Nanobiotechnol. 2022, 20, 360. [Google Scholar] [CrossRef] [PubMed]
  88. Lang, B.T.; Cregg, J.M.; DePaul, M.A.; Tran, A.P.; Xu, K.; Dyck, S.M.; Madalena, K.M.; Brown, B.P.; Weng, Y.-L.; Li, S.; et al. Modulation of the proteoglycan receptor PTPσ promotes recovery after spinal cord injury. Nature 2015, 518, 404–408. [Google Scholar] [CrossRef] [Green Version]
  89. Hu, J.; Shangguan, J.; Askar, P.; Xu, J.; Sun, H.; Zhou, S.; Zhu, C.; Su, W.; Gu, Y. Decellularization alters the unfavorable regenerative adverse microenvironment of the injured spinal cord to support neurite outgrowth. Ann. Transl. Med. 2022, 10, 934. [Google Scholar] [CrossRef]
  90. Cornelison, R.C.; Gonzalez-Rothi, E.J.; Porvasnik, S.L.; Wellman, S.M.; Park, J.H.; Fuller, D.D.; E Schmidt, C. Injectable hydrogels of optimized acellular nerve for injection in the injured spinal cord. Biomed. Mater. 2018, 13, 034110. [Google Scholar] [CrossRef]
  91. Li, C.; Song, J.; Wang, Y.; Shi, Y.; Ji, J.; Lin, Q.; Liu, Y. Adhesion and proliferation of bone marrow stromal cells on acellular spinal cord scaffolds. Int. J. Neurosci. 2022, 1–10. [Google Scholar] [CrossRef]
  92. Baldrick, P. The safety of chitosan as a pharmaceutical excipient. Regul. Toxicol. Pharmacol. 2010, 56, 290–299. [Google Scholar] [CrossRef] [PubMed]
  93. Muzzarelli, R.A.A.; Mattioli-Belmonte, M.; Pugnaloni, A.; Biagini, G. Biochemistry, histology and clinical uses of chitins and chitosans in wound healing. Exs 1999, 87, 251–264. [Google Scholar] [CrossRef] [PubMed]
  94. Gan, Q.; Wang, T. Chitosan nanoparticle as protein delivery carrier—Systematic examination of fabrication conditions for efficient loading and release. Colloids Surfaces B Biointerfaces 2007, 59, 24–34. [Google Scholar] [CrossRef] [PubMed]
  95. Patrulea, V.; Ostafe, V.; Borchard, G.; Jordan, O. Chitosan as a starting material for wound healing applications. Eur. J. Pharm. Biopharm. 2015, 97, 417–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Zuo, Y.Y.; Alolabi, H.; Shafiei, A.; Kang, N.; Policova, Z.; Cox, P.N.; Acosta, E.; Hair, M.L.; Neumann, A.W. Chitosan Enhances the In Vitro Surface Activity of Dilute Lung Surfactant Preparations and Resists Albumin-Induced Inactivation. Pediatr. Res. 2006, 60, 125–130. [Google Scholar] [CrossRef] [Green Version]
  97. Borgens, R.B. Cellular Engineering: Molecular Repair of Membranes to Rescue Cells of the Damaged Nervous System. Neurosurgery 2001, 49, 370–379, discussion 378–379. [Google Scholar] [CrossRef]
  98. Silva, N.A.; Sousa, N.; Reis, R.L.; Salgado, A.J. From basics to clinical: A comprehensive review on spinal cord injury. Prog. Neurobiol. 2014, 114, 25–57. [Google Scholar] [CrossRef]
  99. Luo, J.; Shi, R. Acrolein induces axolemmal disruption, oxidative stress, and mitochondrial impairment in spinal cord tissue. Neurochem. Int. 2004, 44, 475–486. [Google Scholar] [CrossRef]
  100. Tuinstra, H.M.; Aviles, M.O.; Shin, S.; Holland, S.J.; Zelivyanskaya, M.L.; Fast, A.G.; Ko, S.Y.; Margul, D.J.; Bartels, A.K.; Boehler, R.M.; et al. Multifunctional, multichannel bridges that deliver neurotrophin encoding lentivirus for regeneration following spinal cord injury. Biomaterials 2012, 33, 1618–1626. [Google Scholar] [CrossRef] [Green Version]
  101. Gnavi, S.; Barwig, C.; Freier, T.; Haastert-Talini, K.; Grothe, C.; Geuna, S. The Use of Chitosan-Based Scaffolds to Enhance Regeneration in the Nervous System. Int. Rev. Neurobiol. 2013, 109, 1–62. [Google Scholar] [CrossRef]
  102. Nomura, H.; Baladie, B.; Katayama, Y.; Morshead, C.M.; Shoichet, M.S.; Tator, C.H. Delayed implantation of intramedullary chitosan channels containing nerve grafts promotes extensive axonal regeneration after spinal cord injury. Neurosurgery 2008, 63, 127–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Guan, S.; Zhang, X.-L.; Lin, X.-M.; Liu, T.-Q.; Ma, X.-H.; Cui, Z.-F. Chitosan/gelatin porous scaffolds containing hyaluronic acid and heparan sulfate for neural tissue engineering. J. Biomater. Sci. Polym. Ed. 2013, 24, 999–1014. [Google Scholar] [CrossRef] [PubMed]
  104. Kim, M.S.; Kim, G. Three-dimensional electrospun polycaprolactone (PCL)/alginate hybrid composite scaffolds. Carbohydr. Polym. 2014, 114, 213–221. [Google Scholar] [CrossRef] [PubMed]
  105. Chen, S.; Zhao, Y.; Yan, X.; Zhang, L.; Li, G.; Yang, Y. PAM/GO/gel/SA composite hydrogel conduit with bioactivity for repairing peripheral nerve injury. J. Biomed. Mater. Res. Part A 2019, 107, 1273–1283. [Google Scholar] [CrossRef]
  106. Yazdi, M.K.; Taghizadeh, A.; Taghizadeh, M.; Stadler, F.J.; Farokhi, M.; Mottaghitalab, F.; Zarrintaj, P.; Ramsey, J.D.; Seidi, F.; Saeb, M.R.; et al. Agarose-based biomaterials for advanced drug delivery. J. Control. Release 2020, 326, 523–543. [Google Scholar] [CrossRef]
  107. Zarrintaj, P.; Manouchehri, S.; Ahmadi, Z.; Saeb, M.R.; Urbanska, A.M.; Kaplan, D.L.; Mozafari, M. Agarose-based biomaterials for tissue engineering. Carbohydr. Polym. 2018, 187, 66–84. [Google Scholar] [CrossRef]
  108. Balgude, A.P.; Yu, X.; Szymanski, A.; Bellamkonda, R.V. Agarose gel stiffness determines rate of DRG neurite extension in 3D cultures. Biomaterials 2001, 22, 1077–1084. [Google Scholar] [CrossRef]
  109. Luo, Y.; Shoichet, M.S. A photolabile hydrogel for guided three-dimensional cell growth and migration. Nat. Mater. 2004, 3, 249–253. [Google Scholar] [CrossRef]
  110. 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]
  111. Stokols, S.; Sakamoto, J.; Breckon, C.; Holt, T.; Weiss, J.; Tuszynski, M.H. Templated Agarose Scaffolds Support Linear Axonal Regeneration. Tissue Eng. 2006, 12, 2777–2787. [Google Scholar] [CrossRef]
  112. 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] [PubMed]
  113. Gao, M.; Lu, P.; Bednark, B.; Lynam, D.; Conner, J.M.; Sakamoto, J.; Tuszynski, M.H. Templated agarose scaffolds for the support of motor axon regeneration into sites of complete spinal cord transection. Biomaterials 2013, 34, 1529–1536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Lewitus, D.Y.; Smith, K.L.; Landers, J.; Neimark, A.V.; Kohn, J. Bioactive agarose carbon-nanotube composites are capable of manipulating brain-implant interface. J. Appl. Polym. Sci. 2014, 131, 40297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Siddiqi, K.S.; Husen, A.; Sohrab, S.S.; Yassin, M.O. Recent Status of Nanomaterial Fabrication and Their Potential Applications in Neurological Disease Management. Nanoscale Res. Lett. 2018, 13, 231. [Google Scholar] [CrossRef] [PubMed]
  116. Fawcett, J.W. Overcoming Inhibition in the Damaged Spinal Cord. J. Neurotrauma 2006, 23, 371–383. [Google Scholar] [CrossRef]
  117. Bracken, M.B.; Shepard, M.J.; Collins, W.F.; Holford, T.R.; Young, W.; Baskin, D.S.; Eisenberg, H.M.; Flamm, E.; Leo-Summers, L.; Maroon, J.; et al. A Randomized, Controlled Trial of Methylprednisolone or Naloxone in the Treatment of Acute Spinal-Cord Injury: Results of the Second National Acute Spinal Cord Injury Study. N. Engl. J. Med. 1990, 322, 1405–1411. [Google Scholar] [CrossRef]
  118. Gerndt, S.J.; Rodriguez, J.L.; Pawlik, J.W.; Taheri, P.A.; Wahl, W.L.; Micheals, A.J.; Papadopoulos, S.M. Consequences of High-Dose Steroid Therapy for Acute Spinal Cord Injury. J. Trauma Inj. Infect. Crit. Care 1997, 42, 279–284. [Google Scholar] [CrossRef]
  119. Song, F.; Li, X.; Wang, Q.; Liao, L.; Zhang, C. Nanocomposite Hydrogels and Their Applications in Drug Delivery and Tissue Engineering. J. Biomed. Nanotechnol. 2015, 11, 40–52. [Google Scholar] [CrossRef]
  120. Eftekhari, H.; Jahandideh, A.; Asghari, A.; Akbarzadeh, A.; Hesaraki, S. Assessment of polycaprolacton (PCL) nanocomposite scaffold compared with hydroxyapatite (HA) on healing of segmental femur bone defect in rabbits. Artif. Cells Nanomed. Biotechnol. 2017, 45, 961–968. [Google Scholar] [CrossRef]
  121. Huang, X.; Yin, Z.; Wu, S.; Qi, X.; He, Q.; Zhang, Q.; Yan, Q.; Boey, F.; Zhang, H. Graphene-Based Materials: Synthesis, Characterization, Properties, and Applications. Small 2011, 7, 1876–1902. [Google Scholar] [CrossRef]
  122. Wang, X.-X.; Wang, S.-X.; Lu, Y.-B.; Wang, Y.; Song, Y.-J.; Wang, X.; Nyamgerelt, M. Graphene and graphene-based materials in axonal repair of spinal cord injury. Neural Regen. Res. 2022, 17, 2117. [Google Scholar] [CrossRef] [PubMed]
  123. Geim, A.K. Graphene: Status and Prospects. Science 2009, 324, 1530–1534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Dan, Y.; Lu, Y.; Kybert, N.J.; Luo, Z.; Johnson, A.T.C. Intrinsic Response of Graphene Vapor Sensors. Nano Lett. 2009, 9, 1472–1475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Stoller, M.D.; Park, S.; Zhu, Y.; An, J.; Ruoff, R.S. Graphene-Based Ultracapacitors. Nano Lett. 2008, 8, 3498–3502. [Google Scholar] [CrossRef] [PubMed]
  126. Song, Q.; Jiang, Z.; Li, N.; Liu, P.; Liu, L.; Tang, M.; Cheng, G. Anti-inflammatory effects of three-dimensional graphene foams cultured with microglial cells. Biomaterials 2014, 35, 6930–6940. [Google Scholar] [CrossRef]
  127. Ma, Q.; Yang, L.; Jiang, Z.; Song, Q.; Xiao, M.; Zhang, D.; Ma, X.; Wen, T.; Cheng, G. Three-Dimensional Stiff Graphene Scaffold on Neural Stem Cells Behavior. ACS Appl. Mater. Interfaces 2016, 8, 34227–34233. [Google Scholar] [CrossRef]
  128. Sintov, A.; Velasco-Aguirre, C.; Gallardo-Toledo, E.; Araya, E.; Kogan, M. Metal nanoparticles as targeted carriers circumventing the blood–Brain barrier. Int. Rev. Neurobiol. 2016, 130, 199–227. [Google Scholar]
  129. Waris, A.; Ali, A.; Khan, A.U.; Asim, M.; Zamel, D.; Fatima, K.; Raziq, A.; Khan, M.A.; Akbar, N.; Baset, A.; et al. Applications of Various Types of Nanomaterials for the Treatment of Neurological Disorders. Nanomaterials 2022, 12, 2140. [Google Scholar] [CrossRef]
  130. Fang, C.; Ma, Z.; Chen, L.; Li, H.; Jiang, C.; Zhang, W. Biosynthesis of gold nanoparticles, characterization and their loading with zonisamide as a novel drug delivery system for the treatment of acute spinal cord injury. J. Photochem. Photobiol. B Biol. 2019, 190, 72–75. [Google Scholar] [CrossRef]
  131. Wang, Y.-T.; Lu, X.-M.; Zhu, F.; Huang, P.; Yu, Y.; Zeng, L.; Long, Z.-Y.; Wu, Y.-M. The use of a gold nanoparticle-based adjuvant to improve the therapeutic efficacy of hNgR-Fc protein immunization in spinal cord-injured rats. Biomaterials 2011, 32, 7988–7998. [Google Scholar] [CrossRef]
  132. Paviolo, C.; Haycock, J.W.; Yong, J.; Yu, A.; Stoddart, P.R.; McArthur, S.L. Laser exposure of gold nanorods can increase neuronal cell outgrowth. Biotechnol. Bioeng. 2013, 110, 2277–2291. [Google Scholar] [CrossRef] [PubMed]
  133. Naderi, M.S.; Moghadam, T.T.; Khajeh, K.; Ranjbar, B. Improving the stability of chondroitinase ABC I via interaction with gold nanorods. Int. J. Biol. Macromol. 2018, 107, 297–304. [Google Scholar] [CrossRef] [PubMed]
  134. Lin, F.; Liu, Y.; Luo, W.; Liu, S.; Wang, Y.; Gu, R.; Liu, W.; Xiao, C. Minocycline-Loaded Poly(α-Lipoic Acid)–Methylprednisolone Prodrug Nanoparticles for the Combined Anti-Inflammatory Treatment of Spinal Cord Injury. Int. J. Nanomed. 2022, 17, 91–104. [Google Scholar] [CrossRef]
  135. Shi, H.; Jin, L.; Li, J.; Liang, K.; Li, X.; Ye, Z.; Zhu, X.; Oliveira, J.M.; Reis, R.L.; Mao, Z.; et al. Mesoporous polydopamine nanoparticles for sustained release of rapamycin and reactive oxygen species scavenging to synergistically accelerate neurogenesis after spinal cord injury. J. Mater. Chem. B 2022, 10, 6351–6359. [Google Scholar] [CrossRef] [PubMed]
  136. Lee, S.; Trinh, T.H.; Yoo, M.; Shin, J.; Lee, H.; Kim, J.; Hwang, E.; Lim, Y.-B.; Ryou, C. Self-Assembling Peptides and Their Application in the Treatment of Diseases. Int. J. Mol. Sci. 2019, 20, 5850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. La Manna, S.; Di Natale, C.; Florio, D.; Marasco, D. Peptides as Therapeutic Agents for Inflammatory-Related Diseases. Int. J. Mol. Sci. 2018, 19, 2714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Guo, J.; Leung, G.K.K.; Su, H.; Yuan, Q.; Wang, L.; Chu, T.-H.; Zhang, W.; Pu, J.K.S.; Ng, G.K.P.; Wong, W.M.; et al. Self-assembling peptide nanofiber scaffold promotes the reconstruction of acutely injured brain. Nanomedicine 2009, 5, 345–351. [Google Scholar] [CrossRef] [PubMed]
  139. Cheng, T.-Y.; Chen, M.-H.; Chang, W.-H.; Huang, M.-Y.; Wang, T.-W. Neural stem cells encapsulated in a functionalized self-assembling peptide hydrogel for brain tissue engineering. Biomaterials 2013, 34, 2005–2016. [Google Scholar] [CrossRef]
  140. Ando, K.; Imagama, S.; Ito, Z.; Kobayashi, K.; Hida, T.; Nakashima, H.; Ito, K.; Tsushima, M.; Ishikawa, Y.; Matsumoto, A.; et al. Self-assembling Peptide Reduces Glial Scarring, Attenuates Posttraumatic Inflammation, and Promotes Neurite Outgrowth of Spinal Motor Neurons. Spine 2016, 41, E1201–E1207. [Google Scholar] [CrossRef]
  141. Subramanian, A.; Krishnan, U.M.; Sethuraman, S. Development of biomaterial scaffold for nerve tissue engineering: Biomaterial mediated neural regeneration. J. Biomed. Sci. 2009, 16, 108. [Google Scholar] [CrossRef] [Green Version]
  142. Ratheesh, G.; Venugopal, J.R.; Chinappan, A.; Ezhilarasu, H.; Sadiq, A.; Ramakrishna, S. 3D Fabrication of Polymeric Scaffolds for Regenerative Therapy. ACS Biomater. Sci. Eng. 2017, 3, 1175–1194. [Google Scholar] [CrossRef] [PubMed]
  143. Liu, H.; Slamovich, E.B.; Webster, T.J. Less harmful acidic degradation of poly(lactic-co-glycolic acid) bone tissue engineering scaffolds through titania nanoparticle addition. Int. J. Nanomed. 2006, 1, 541–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Shalumon, K.T.; Anulekha, K.H.; Nair, S.V.; Nair, S.V.; Chennazhi, K.P.; Jayakumar, R. Sodium alginate/poly(vinyl alcohol)/nano ZnO composite nanofibers for antibacterial wound dressings. Int. J. Biol. Macromol. 2011, 49, 247–254. [Google Scholar] [CrossRef] [PubMed]
  145. Gunatillake, P.A.; Adhikari, R. Biodegradable synthetic polymers for tissue engineering. Eur. Cells Mater. 2003, 5, 1–16, discussion 16. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, P.; Wang, H.; Ma, K.; Wang, S.; Yang, C.; Mu, N.; Yang, F.; Feng, H.; Chen, T. Novel cytokine-loaded PCL-PEG scaffold composites for spinal cord injury repair. RSC Adv. 2020, 10, 6306–6314. [Google Scholar] [CrossRef]
  147. Luo, J.; Shi, R. Diffusive oxidative stress following acute spinal cord injury in guinea pigs and its inhibition by polyethylene glycol. Neurosci. Lett. 2004, 359, 167–170. [Google Scholar] [CrossRef]
  148. Shi, R. Polyethylene glycol repairs membrane damage and enhances functional recovery: A tissue engineering approach to spinal cord injury. Neurosci. Bull. 2013, 29, 460–466. [Google Scholar] [CrossRef] [Green Version]
  149. Wang, A.; Huo, X.; Zhang, G.; Wang, X.; Zhang, C.; Wu, C.; Rong, W.; Xu, J.; Song, T. Effect of DSPE-PEG on compound action potential, injury potential and ion concentration following compression in ex vivo spinal cord. Neurosci. Lett. 2016, 620, 50–56. [Google Scholar] [CrossRef]
  150. Raynald; Shu, B.; Liu, X.; Zhou, J.; Huang, H.; Wang, J.; Sun, X.; Qin, C.; An, Y. 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]
  151. Liu, Z.-H.; Huang, Y.-C.; Kuo, C.-Y.; Kuo, C.-Y.; Chin, C.-Y.; Yip, P.K.; Chen, J.-P. Docosahexaenoic Acid-Loaded Polylactic Acid Core-Shell Nanofiber Membranes for Regenerative Medicine after Spinal Cord Injury: In Vitro and In Vivo Study. Int. J. Mol. Sci. 2020, 21, 7031. [Google Scholar] [CrossRef]
  152. Hurtado, A.; Cregg, J.; Wang, H.B.; Wendell, D.F.; Oudega, M.; Gilbert, R.J.; McDonald, J.W. Robust CNS regeneration after complete spinal cord transection using aligned poly-l-lactic acid microfibers. Biomaterials 2011, 32, 6068–6079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Gwak, S.-J.; Yun, Y.; Yoon, D.H.; Kim, K.N.; Ha, Y. Therapeutic Use of 3β-[N-(N′,N′-Dimethylaminoethane) Carbamoyl] Cholesterol-Modified PLGA Nanospheres as Gene Delivery Vehicles for Spinal Cord Injury. PLoS ONE 2016, 11, e0147389. [Google Scholar] [CrossRef] [PubMed]
  154. Ganz, J.; Shor, E.; Guo, S.; Sheinin, A.; Arie, I.; Michaelevski, I.; Pitaru, S.; Offen, D.; Levenberg, S. Implantation of 3D Constructs Embedded with Oral Mucosa-Derived Cells Induces Functional Recovery in Rats with Complete Spinal Cord Transection. Front. Neurosci. 2017, 11, 589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Guo, S.; Redenski, I.; Landau, S.; Szklanny, A.; Merdler, U.; Levenberg, S. Prevascularized Scaffolds Bearing Human Dental Pulp Stem Cells for Treating Complete Spinal Cord Injury. Adv. Health Mater. 2020, 9, e2000974. [Google Scholar] [CrossRef] [PubMed]
  156. 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]
  157. Babaloo, H.; Ebrahimi-Barough, S.; Derakhshan, M.A.; Yazdankhah, M.; Lotfibakhshaiesh, N.; Soleimani, M.; Joghataei, M.; 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]
  158. 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]
  159. Shahriari, D.; Koffler, J.Y.; Tuszynski, M.H.; Campana, W.M.; Sakamoto, J.S. Hierarchically Ordered Porous and High-Volume Polycaprolactone Microchannel Scaffolds Enhanced Axon Growth in Transected Spinal Cords. Tissue Eng. Part A 2017, 23, 415–425. [Google Scholar] [CrossRef] [Green Version]
  160. Comolli, N.; Donaldson, O.; Grantier, N.; Zhukareva, V.; Tom, V.J. Polyvinyl alcohol-polyvinyl pyrrolidone thin films provide local short-term release of anti-inflammatory agents post spinal cord injury. J. Biomed. Mater. Res. Part B Appl. Biomater. 2012, 100, 1867–1873. [Google Scholar] [CrossRef]
  161. Chen, L.; Wang, W.; Lin, Z.; Lu, Y.; Chen, H.; Li, B.; Li, Z.; Xia, H.; Li, L.; Zhang, T. Conducting molybdenum sulfide/graphene oxide/polyvinyl alcohol nanocomposite hydrogel for repairing spinal cord injury. J. Nanobiotechnol. 2022, 20, 210. [Google Scholar] [CrossRef]
  162. 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]
  163. Zhao, T.; Xu, K.; Wu, Q.; Wang, C.; Xiao, S.; Li, H.; He, T.; Wang, L.; Li, F.; Chen, Q. Duraplasty of PHBV/PLA/Col membranes promotes axonal regeneration by inhibiting NLRP3 complex and M1 macrophage polarization in rats with spinal cord injury. FASEB J. 2020, 34, 12147–12162. [Google Scholar] [CrossRef] [PubMed]
  164. Kong, G.; Liu, J.; Li, R.; Lin, J.; Huang, Z.; Yang, Z.; Wu, X.; Huang, Z.; Zhu, Q.; Wu, X. Ketone Metabolite β-Hydroxybutyrate Ameliorates Inflammation After Spinal Cord Injury by Inhibiting the NLRP3 Inflammasome. Neurochem. Res. 2021, 46, 213–229. [Google Scholar] [CrossRef]
  165. Zhang, S.; Wang, X.-J.; Li, W.-S.; Xu, X.-L.; Hu, J.-B.; Kang, X.-Q.; Qi, J.; Ying, X.-Y.; You, J.; Du, Y.-Z. Polycaprolactone/polysialic acid hybrid, multifunctional nanofiber scaffolds for treatment of spinal cord injury. Acta Biomater. 2018, 77, 15–27. [Google Scholar] [CrossRef]
  166. Mehanna, A.; Jakovcevski, I.; Acar, A.; Xiao, M.; Loers, G.; Rougon, G.; Irintchev, A.; Schachner, M. Polysialic Acid Glycomimetic Promotes Functional Recovery and Plasticity After Spinal Cord Injury in Mice. Mol. Ther. 2010, 18, 34–43. [Google Scholar] [CrossRef]
  167. 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. New York Acad. Sci. 2020, 1465, 99–116. [Google Scholar] [CrossRef]
  168. Tsai, E.; Dalton, P.D.; Shoichet, M.S.; Tator, C.H. Matrix inclusion within synthetic hydrogel guidance channels improves specific supraspinal and local axonal regeneration after complete spinal cord transection. Biomaterials 2006, 27, 519–533. [Google Scholar] [CrossRef]
  169. 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. 2013, 9, 1298–1309. [Google Scholar] [CrossRef]
  170. Tsai, E.; Dalton, P.D.; Shoichet, M.S.; Tator, C.H. Synthetic Hydrogel Guidance Channels Facilitate Regeneration of Adult Rat Brainstem Motor Axons after Complete Spinal Cord Transection. J. Neurotrauma 2004, 21, 789–804. [Google Scholar] [CrossRef]
  171. D’Souza, A.A.; Shegokar, R. Polyethylene glycol (PEG): A versatile polymer for pharmaceutical applications. Expert Opin. Drug Deliv. 2016, 13, 1257–1275. [Google Scholar] [CrossRef]
  172. Pape, A.C.H.; Ippel, B.D.; Dankers, P.Y.W. Cell and Protein Fouling Properties of Polymeric Mixtures Containing Supramolecular Poly(ethylene glycol) Additives. Langmuir 2017, 33, 4076–4082. [Google Scholar] [CrossRef] [PubMed]
  173. Knop, K.; Hoogenboom, R.; Fischer, D.; Schubert, U.S. Poly(ethylene glycol) in Drug Delivery: Pros and Cons as Well as Potential Alternatives. Angew. Chem. Int. Ed. 2010, 49, 6288–6308. [Google Scholar] [CrossRef] [PubMed]
  174. Padín-González, E.; Lancaster, P.; Bottini, M.; Gasco, P.; Tran, L.; Fadeel, B.; Wilkins, T.; Monopoli, M.P. Understanding the Role and Impact of Poly (Ethylene Glycol) (PEG) on Nanoparticle Formulation: Implications for COVID-19 Vaccines. Front. Bioeng. Biotechnol. 2022, 10, 882363. [Google Scholar] [CrossRef] [PubMed]
  175. Tyler, J.Y.; Xu, X.-M.; Cheng, J.-X. Nanomedicine for treating spinal cord injury. Nanoscale 2013, 5, 8821–8836. [Google Scholar] [CrossRef] [Green Version]
  176. Carballo-Molina, O.A.; Velasco, I. Hydrogels as scaffolds and delivery systems to enhance axonal regeneration after injuries. Front. Cell. Neurosci. 2015, 9, 13. [Google Scholar] [CrossRef] [Green Version]
  177. Peppas, N.A.; Bures, P.; Leobandung, W.; Ichikawa, H. Hydrogels in pharmaceutical formulations. Eur. J. Pharm. Biopharm. 2000, 50, 27–46. [Google Scholar] [CrossRef]
  178. Rizzi, S.C.; Ehrbar, M.; Halstenberg, S.; Raeber, G.P.; Schmoekel, H.G.; Hagenmüller, H.; Müller, R.; Weber, F.E.; Hubbell, J.A. Recombinant Protein-co-PEG Networks as Cell-Adhesive and Proteolytically Degradable Hydrogel Matrixes. Part II: Biofunctional Characteristics. Biomacromolecules 2006, 7, 3019–3029. [Google Scholar] [CrossRef]
  179. Benoit, D.; Schwartz, M.; Durney, A.R.; Anseth, K.S. Small functional groups for controlled differentiation of hydrogel-encapsulated human mesenchymal stem cells. Nat. Mater. 2008, 7, 816–823. [Google Scholar] [CrossRef] [Green Version]
  180. Luo, J.; Borgens, R.; Shi, R. Polyethylene glycol immediately repairs neuronal membranes and inhibits free radical production after acute spinal cord injury. J. Neurochem. 2002, 83, 471–480. [Google Scholar] [CrossRef]
  181. Lopes, M.S.; Jardini, A.; Filho, R.M. Poly (Lactic Acid) Production for Tissue Engineering Applications. Procedia Eng. 2012, 42, 1402–1413. [Google Scholar] [CrossRef] [Green Version]
  182. Gupta, B.; Revagade, N.; Hilborn, J. Poly(lactic acid) fiber: An overview. Prog. Polym. Sci. 2007, 32, 455–482. [Google Scholar] [CrossRef]
  183. Gautier, S.E.; Oudega, M.; Fragoso, M.; Chapon, P.; Plant, G.W.; Bunge, M.B.; Parel, J.M. Poly(alpha-hydroxyacids) for application in the spinal cord: Resorbability and biocompatibility with adult rat Schwann cells and spinal cord. J. Biomed. Mater. Res. 1998, 42, 642–654. [Google Scholar] [CrossRef]
  184. Lasprilla, A.J.R.; Martinez, G.A.R.; Lunelli, B.H.; Jardini, A.L.; Filho, R.M. Poly-lactic acid synthesis for application in biomedical devices—A review. Biotechnol. Adv. 2012, 30, 321–328. [Google Scholar] [CrossRef] [PubMed]
  185. Rasal, R.M.; Janorkar, A.V.; Hirt, D.E. Poly(lactic acid) modifications. Prog. Polym. Sci. 2010, 35, 338–356. [Google Scholar] [CrossRef]
  186. Burg, K.J.L.; Holder, W.D.; Culberson, C.R.; Beiler, R.J.; Greene, K.G.; Loebsack, A.B.; Roland, W.D.; Mooney, D.J.; Halberstadt, C.R. Parameters affecting cellular adhesion to polylactide films. J. Biomater. Sci. Polym. Ed. 1999, 10, 147–161. [Google Scholar] [CrossRef] [PubMed]
  187. Rasal, R.M.; Hirt, D.E. Toughness decrease of PLA-PHBHHx blend films upon surface-confined photopolymerization. J. Biomed. Mater. Res. Part A 2009, 88, 1079–1086. [Google Scholar] [CrossRef] [PubMed]
  188. Danhier, F.; Ansorena, E.; Silva, J.M.; Coco, R.; Le Breton, A.; Préat, V. PLGA-based nanoparticles: An overview of biomedical applications. J. Control. Release 2012, 161, 505–522. [Google Scholar] [CrossRef]
  189. Rocha, C.V.; Gonçalves, V.; da Silva, M.C.; Bañobre-López, M.; Gallo, J. PLGA-Based Composites for Various Biomedical Applications. Int. J. Mol. Sci. 2022, 23, 2034. [Google Scholar] [CrossRef]
  190. Mirakabad, F.S.T.; Nejati-Koshki, K.; Akbarzadeh, A.; Yamchi, M.R.; Milani, M.; Zarghami, N.; Zeighamian, V.; Rahimzadeh, A.; Alimohammadi, S.; Hanifehpour, Y.; et al. PLGA-Based Nanoparticles as Cancer Drug Delivery Systems. Asian Pac. J. Cancer Prev. 2014, 15, 517–535. [Google Scholar] [CrossRef]
  191. Makadia, H.K.; Siegel, S.J. Poly lactic-co-glycolic acid (PLGA) As biodegradable controlled drug delivery carrier. Polymers 2011, 3, 1377–1397. [Google Scholar] [CrossRef]
  192. Han, F.Y.; Thurecht, K.J.; Whittaker, A.K.; Smith, M.T. Bioerodable PLGA-Based Microparticles for Producing Sustained-Release Drug Formulations and Strategies for Improving Drug Loading. Front. Pharmacol. 2016, 7, 185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Ding, D.; Zhu, Q. Recent advances of PLGA micro/nanoparticles for the delivery of biomacromolecular therapeutics. Mater. Sci. Eng. C 2018, 92, 1041–1060. [Google Scholar] [CrossRef] [PubMed]
  194. Krych, A.J.; Rooney, G.E.; Chen, B.; Schermerhorn, T.C.; Ameenuddin, S.; Gross, L.; Moore, M.J.; Currier, B.L.; Spinner, R.J.; Friedman, J.A.; et al. Relationship between scaffold channel diameter and number of regenerating axons in the transected rat spinal cord. Acta Biomater. 2009, 5, 2551–2559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Mohtaram, N.K.; Ko, J.; King, C.; Sun, L.; Muller, N.; Jun, M.B.-G.; Willerth, S.M. Electrospun biomaterial scaffolds with varied topographies for neuronal differentiation of human-induced pluripotent stem cells. J. Biomed. Mater. Res. Part A 2014, 103, 2591–2601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. He, L.; Zhang, Y.; Zeng, C.; Ngiam, M.; Liao, S.; Quan, D.; Zeng, Y.; Lu, J.; Ramakrishna, S. Manufacture of PLGA Multiple-Channel Conduits with Precise Hierarchical Pore Architectures and In Vitro/Vivo Evaluation for Spinal Cord Injury. Tissue Eng. Part C Methods 2009, 15, 243–255. [Google Scholar] [CrossRef]
  197. Sonomoto, K.; Yamaoka, K.; Kaneko, H.; Yamagata, K.; Sakata, K.; Zhang, X.; Kondo, M.; Zenke, Y.; Sabanai, K.; Nakayamada, S.; et al. Spontaneous Differentiation of Human Mesenchymal Stem Cells on Poly-Lactic-Co-Glycolic Acid Nano-Fiber Scaffold. PLoS ONE 2016, 11, e0153231. [Google Scholar] [CrossRef] [Green Version]
  198. 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]
  199. Liu, C.; Huang, Y.; Pang, M.; Yang, Y.; Linshan, L.; Liu, L.; Shu, T.; Zhou, W.; Wang, X.; Rong, L.; et al. Tissue-Engineered Regeneration of Completely Transected Spinal Cord Using Induced Neural Stem Cells and Gelatin-Electrospun Poly (Lactide-Co-Glycolide)/Polyethylene Glycol Scaffolds. PLoS ONE 2015, 10, e0117709. [Google Scholar] [CrossRef] [Green Version]
  200. Sawadkar, P.; Mohanakrishnan, J.; Rajasekar, P.; Rahmani, B.; Kohli, N.; Bozec, L.; Garcia-Gareta, E. A Synergistic Relationship between Polycaprolactone and Natural Polymers Enhances the Physical Properties and Biological Activity of Scaffolds. ACS Appl. Mater. Interfaces 2020, 12, 13587–13597. [Google Scholar] [CrossRef]
  201. Lim, W.; Chowdhury, S.; Ng, M.; Law, J. Physicochemical Properties and Biocompatibility of Electrospun Polycaprolactone/Gelatin Nanofibers. Int. J. Environ. Res. Public Health 2021, 18, 4764. [Google Scholar] [CrossRef]
  202. Patel, B.B.; Sharifi, F.; Stroud, D.P.; Montazami, R.; Hashemi, N.N.; Sakaguchi, D.S. 3D Microfibrous Scaffolds Selectively Promotes Proliferation and Glial Differentiation of Adult Neural Stem Cells: A Platform to Tune Cellular Behavior in Neural Tissue Engineering. Macromol. Biosci. 2019, 19, e1800236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. 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]
  204. Hiraizumi, Y.; Transfeldt, E.E.; Fujimaki, E.; Nambu, M. Application of Polyvinyl Alcohol Hydrogel Membrane as Anti-adhesive Interposition After Spinal Surgery. Spine 1995, 20, 2272–2277. [Google Scholar] [CrossRef] [PubMed]
  205. Lebugle, A.; Rodrigues, A.; Bonnevialle, P.; Voigt, J.; Canal, P.; Rodriguez, F. Study of implantable calcium phosphate systems for the slow release of methotrexate. Biomaterials 2002, 23, 3517–3522. [Google Scholar] [CrossRef]
  206. 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]
  207. Doncel-Pérez, E.; Ellis, G.; Sandt, C.; Shuttleworth, P.S.; Bastida, A.; Revuelta, J.; García-Junceda, E.; Fernández-Mayoralas, A.; Garrido, L. Biochemical profiling of rat embryonic stem cells grown on electrospun polyester fibers using synchrotron infrared microspectroscopy. Anal. Bioanal. Chem. 2018, 410, 3649–3660. [Google Scholar] [CrossRef] [Green Version]
  208. Agrawal, L.; Vimal, S.K.; Barzaghi, P.; Shiga, T.; Terenzio, M. Biodegradable and Electrically Conductive Melanin-Poly (3-Hydroxybutyrate) 3D Fibrous Scaffolds for Neural Tissue Engineering Applications. Macromol. Biosci. 2022, 22, e2200315. [Google Scholar] [CrossRef]
  209. Wu, J.; Zhan, X.; Liu, L.; Xia, X. Bioproduction, purification, and application of polysialic acid. Appl. Microbiol. Biotechnol. 2018, 102, 9403–9409. [Google Scholar] [CrossRef]
  210. Mindler, K.; Ostertag, E.; Stehle, T. The polyfunctional polysialic acid: A structural view. Carbohydr. Res. 2021, 507, 108376. [Google Scholar] [CrossRef]
  211. Pearse, D.D.; Rao, S.N.; Morales, A.A.; Wakarchuk, W.; Rutishauser, U.; El-Maarouf, A.; Ghosh, M. Engineering polysialic acid on Schwann cells using polysialyltransferase gene transfer or purified enzyme exposure for spinal cord injury transplantation. Neurosci. Lett. 2021, 748, 135690. [Google Scholar] [CrossRef]
  212. Wang, X.-J.; Peng, C.-H.; Zhang, S.; Xu, X.-L.; Shu, G.-F.; Qi, J.; Zhu, Y.-F.; Xu, D.-M.; Kang, X.-Q.; Lu, K.-J.; et al. Polysialic-Acid-Based Micelles Promote Neural Regeneration in Spinal Cord Injury Therapy. Nano Lett. 2019, 19, 829–838. [Google Scholar] [CrossRef] [PubMed]
  213. Marino, P.; Norreel, J.-C.; Schachner, M.; Rougon, G.; Amoureux, M.-C. A polysialic acid mimetic peptide promotes functional recovery in a mouse model of spinal cord injury. Exp. Neurol. 2009, 219, 163–174. [Google Scholar] [CrossRef] [PubMed]
  214. Giannetti, S.; Lauretti, L.; Fernandez, E.; Salvinelli, F.; Tamburrini, G.; Pallini, R. Acrylic hydrogel implants after spinal cord lesion in the adult rat. Neurol. Res. 2001, 23, 405–409. [Google Scholar] [CrossRef]
  215. 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] [PubMed]
  216. Hejčl, A.; Lesný, P.; Přádný, M.; Šedý, J.; Zámečník, J.; Jendelová, P.; Michálek, J.; Syková, E. Macroporous hydrogels based on 2-hydroxyethyl methacrylate. Part 6: 3D hydrogels with positive and negative surface charges and polyelectrolyte complexes in spinal cord injury repair. J. Mater. Sci. Mater. Med. 2009, 20, 1571–1577. [Google Scholar] [CrossRef] [PubMed]
  217. Kubinová, Š.; Horák, D.; Vaněček, V.; Plichta, Z.; Proks, V.; Syková, E. The use of new surface-modified poly(2-hydroxyethyl methacrylate) hydrogels in tissue engineering: Treatment of the surface with fibronectin subunits versus Ac-CGGASIKVAVS-OH, cysteine, and 2-mercaptoethanol modification. J. Biomed. Mater. Res. Part A 2013, 102, 2315–2323. [Google Scholar] [CrossRef] [PubMed]
  218. Carone, T.W.; Hasenwinkel, J.M. Mechanical and morphological characterization of homogeneous and bilayered poly(2-hydroxyethyl methacrylate) scaffolds for use in CNS nerve regeneration. J. Biomed. Mater. Res. Part B Appl. Biomater. 2006, 78, 274–282. [Google Scholar] [CrossRef] [PubMed]
  219. Valdes-Sánchez, T.; Rodriguez-Jimenez, F.J.; García-Cruz, D.M.; Escobar-Ivirico, J.L.; Alastrue-Agudo, A.; Erceg, S.; Monleón, M.; Moreno-Manzano, V. Methacrylate-endcapped caprolactone and FM19G11 provide a proper niche for spinal cord-derived neural cells. J. Tissue Eng. Regen. Med. 2013, 9, 734–739. [Google Scholar] [CrossRef]
  220. Hejčl, A.; Růžička, J.; Proks, V.; Macková, H.; Kubinová, Š.; Tukmachev, D.; Cihlář, J.; Horák, D.; Jendelová, P. Dynamics of tissue ingrowth in SIKVAV-modified highly superporous PHEMA scaffolds with oriented pores after bridging a spinal cord transection. J. Mater. Sci. Mater. Med. 2018, 29, 89. [Google Scholar] [CrossRef]
  221. Geetha, M.; Singh, A.K.; Asokamani, R.; Gogia, A.K. Ti based biomaterials, the ultimate choice for orthopaedic implants—A review. Prog. Mater. Sci. 2009, 54, 397–425. [Google Scholar] [CrossRef]
  222. Chen, Q.; Thouas, G.A. Metallic implant biomaterials. Mater. Sci. Eng. R Rep. 2015, 87, 1–57. [Google Scholar] [CrossRef]
  223. Spataru, M.-C.; Cojocaru, F.D.; Sandu, A.V.; Solcan, C.; Duceac, I.A.; Baltatu, M.S.; Voiculescu, I.; Geanta, V.; Vizureanu, P. Assessment of the Effects of Si Addition to a New TiMoZrTa System. Materials 2021, 14, 7610. [Google Scholar] [CrossRef] [PubMed]
  224. Che Lah, N.A.; Hussin, M.H. Titanium and Titanium Based Alloys as Metallic Biomaterials in Medical Applications-Spine Implant Case Study. Pertanika J. Sci. Technol. 2019, 27, 459–472. [Google Scholar]
  225. 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]
  226. Wang, J.; Kong, X.; Li, Q.; Li, C.; Yu, H.; Ning, G.; Xiang, Z.; Liu, Y.; Feng, S. The spatial arrangement of cells in a 3D-printed biomimetic spinal cord promotes directional differentiation and repairs the motor function after spinal cord injury. Biofabrication 2021, 13, 045016. [Google Scholar] [CrossRef] [PubMed]
  227. Liu, X.-Y.; Chen, C.; Xu, H.-H.; Zhang, Y.-S.; Zhong, L.; Hu, N.; Jia, X.-L.; Wang, Y.-W.; Zhong, K.-H.; Liu, C.; et al. Integrated printed BDNF/collagen/chitosan scaffolds with low temperature extrusion 3D printer accelerated neural regeneration after spinal cord injury. Regen. Biomater. 2021, 8, rbab047. [Google Scholar] [CrossRef] [PubMed]
  228. Li, X.-H.; Zhu, X.; Liu, X.-Y.; Xu, H.-H.; Jiang, W.; Wang, J.-J.; Chen, F.; Zhang, S.; Li, R.-X.; Chen, X.-Y.; et al. The corticospinal tract structure of collagen/silk fibroin scaffold implants using 3D printing promotes functional recovery after complete spinal cord transection in rats. J. Mater. Sci. Mater. Med. 2021, 32, 31. [Google Scholar] [CrossRef]
  229. Joung, D.; Truong, V.; Neitzke, C.C.; Guo, S.-Z.; Walsh, P.J.; Monat, J.R.; Meng, F.; Park, S.H.; Dutton, J.R.; Parr, A.M.; et al. 3D Printed Stem-Cell Derived Neural Progenitors Generate Spinal Cord Scaffolds. Adv. Funct. Mater. 2018, 28. [Google Scholar] [CrossRef]
  230. 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]
  231. 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]
  232. Xiao, Z.; Tang, F.; Tang, J.; Yang, H.; Zhao, Y.; Chen, B.; Han, S.; Wang, N.; Li, X.; Cheng, S.; et al. One-year clinical study of NeuroRegen scaffold implantation following scar resection in complete chronic spinal cord injury patients. Sci. China Life Sci. 2016, 59, 647–655. [Google Scholar] [CrossRef] [PubMed]
  233. 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] [PubMed] [Green Version]
  234. 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]
  235. Theodore, N.; Hlubek, R.; Danielson, J.; Neff, K.; Vaickus, L.; Ulich, T.R.; Ropper, A.E. First Human Implantation of a Bioresorbable Polymer Scaffold for Acute Traumatic Spinal Cord Injury: A Clinical Pilot Study for Safety and Feasibility. Neurosurgery 2016, 79, E305–E312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Chen, X.-Y.; Zhang, S.; Deng, W.-S.; Ma, K.; Liang, B.; Liu, X.-Y.; Xu, H.-Y.; Zhang, J.; Shi, H.-Y.; Sun, H.-T. Collagen scaffold combined with human umbilical cord-mesenchymal stem cells transplantation for acute complete spinal cord injury. Neural Regen. Res. 2020, 15, 1686–1700. [Google Scholar] [CrossRef]
Figure 1. The pathological mechanism in the injury site after SCI. (A). Inflammatory cell infiltration and the release of neurotoxins were generated from the inflammatory cascade response of immune cells at the acute stage of SCI, which impeded axonal regeneration in the injury site of SCI. (B). Cystic cavities, myelin debris, deposition of GSPGs and astrocytes were progressively generated at the chronic stage of SCI.
Figure 1. The pathological mechanism in the injury site after SCI. (A). Inflammatory cell infiltration and the release of neurotoxins were generated from the inflammatory cascade response of immune cells at the acute stage of SCI, which impeded axonal regeneration in the injury site of SCI. (B). Cystic cavities, myelin debris, deposition of GSPGs and astrocytes were progressively generated at the chronic stage of SCI.
Ijms 24 00816 g001
Figure 2. Current treatments for SCI include surgery, drug-targeted therapy, biomaterials, stem cell therapy, and tissue engineering scaffolds.
Figure 2. Current treatments for SCI include surgery, drug-targeted therapy, biomaterials, stem cell therapy, and tissue engineering scaffolds.
Ijms 24 00816 g002
Figure 3. Characteristics and classification of biomaterials.
Figure 3. Characteristics and classification of biomaterials.
Ijms 24 00816 g003
Table 1. The bioactivity of natural biomaterials in various models of SCI.
Table 1. The bioactivity of natural biomaterials in various models of SCI.
Natural BiomaterialsImplanted SubstanceAnimals and SCI ModelEffectRef.
Hyaluronic AcidHuman embryonic stem cell-derived neural stem cellsWister rats with spinal cord injuryIncreasing cell differentiation and improving motor function[35]
Hyaluronic AcidNogo-66 receptor antibody and poly-L-lysineWister rats with spinal cord hemisectionPromoting vascular regeneration, and inhibiting the formation of glial scar[36]
CollagenLinear ordered collagen scaffolds loaded with collagen-binding neurotrophin-3Wister rats with spinal cord transectionPromoting axonal regeneration and restoring some motor function[37]
CollagenNeurotrophin-3Dawley rats with spinal cord hemisectionInhibiting inflammation and scar; promoting neuronal regeneration[38]
CollagenImplanted vascular endothelial growth factorSprague–Dawley (SD) rats with spinal cord transectionImproving the microenvironment and promoting angiogenesis[39]
CollagenN-cadherinSD rats with spinal cord transectionPromoting the regeneration of neurons and improving motor function[40]
CollagenHuman placenta-derived mesenchymal stem cellsBeagle dog with spinal cord transectionImproving the regeneration of neurons and exercise capacity[41]
CollagenFreeze-dried alginate sponge cross-linked with covalent bondsRats with spinal cord transectionEnhancing nerve regeneration of spinal cord[42]
GelatiniNSCsC57BL/6N mice with spinal cord transectionPromoting the proliferation of neural stem cells and significantly promoting functional recovery[43]
GelatinBMSC and NSCSD rats with spinal cord hemisectionSignificantly promoting motor function recovery and neuronal differentiation[44]
GelatinNSCSD rats with spinal cord transectionSignificantly improving hindlimb movement and nerve regeneration[45]
FibrinPot-bellied pigs with spinal cord dorsal column removedPromoting tissue repair in areas near the injury and restoration of conduction function in the posterior column of the spinal cord[46]
FibrinSD rats with spinal cord hemisectionPromoting directed host cell invasion, vasculature remodeling and axonal regeneration[47]
FibrinHuman endometrial stem cells (hEnSCs)Wister rats with spinal cord aneurysm clipSignificantly promoting the recovery of motor function in injured rats[48]
Decellularized scaffoldiPN and SCFischer rats with Moderate thoracic contusion injuryPromoting Schwann cell survival and neurite outgrowth in grafts[49]
Decellularized scaffoldbpV (pic)SD rats with spinal cord hemisectionPromoting NSC activity and axon outgrowth[50]
Decellularized scaffoldADSCSD rats with spinal cord hemisecionEnhancing the ability of axonal regeneration and promoting functional recovery[51]
ChitosanWister rats with spinal cord hemisectionReducing glial scar; improving the inflammatory response[52]
ChitosanNeurotrophin-3 and human umbilical cord mesenchymal stem cellsC57BL/6 Mice with spinal cord transectionPromoting the recovery of neurological function and reducing inflammation[53]
AlginateNeural stem cellsSD rats with spinal cord Aneurysmal clipReducing inflammation and lesion size[54]
AlginatePeptides and astrogliaGFP-transgenic F344 rats with spinal cord hemisectionPromoting cell migration and slight axon growth[55]
AgaroseBDNFF344 rats with spinal cord transectionPromoting nerve regeneration and axon growth[56]
AgaroseMatrigelSprague–Dawley rats with spinal cord dorsal column removedImproving motor function and promoting cell proliferation[57]
AgaroseGelatin and polypyrroleRats with spinal cord hemisectionInhibiting the formation of astrocytes and activating endogenous nerve regeneration of spinal cord[58]
NanomaterialMPSD rats with spinal cord hemisectionReducing lesion volume and improving drug transport efficiency[59]
NanomaterialMPWister rats with spinal cord hemisectionReducing the secondary reaction after SCI[60]
NanomaterialPoly(lactide-coglycolide)C57/BL6 mice with spinal cord hemisectionEnhancing the expression of anti-inflammatory and regeneration genes and increasing axon regeneration[61]
NanomaterialChitosan and polyethylene glycolBALB/c mice with compression injury of spinal cordPromoting cell growth and reducing inflammatory response[62]
Self-loading peptideNeural precursor cellsWister rats with spinal cord aneurysm clipEnhancing nerve repair and regeneration[63]
Self-loading peptideSprague–Dawley rats with thoracic spinal cord tissuesPromoting the proliferation and migration of neural stem cells[64]
Self-loading peptideSD rats with injury spinal cordReducing inflammation and glial scar formation, and increaseing axonal growth[65]
–: There is no implanted substance
Table 2. The bioactivity of synthetic biomaterials in various models of SCI.
Table 2. The bioactivity of synthetic biomaterials in various models of SCI.
Synthetic BiomaterialsImplantted SubstanceAnimals and SCI ModelEffectRef.
PEGFGF2, EGF, GDNFSD Rats with spinal cord transectionImproving motor function and increasing axonal regeneration[146]
PEGGuinea pig with compression injury of spinal cordInhibiting the formation of free radicals, and resistancing lipid peroxidation[147]
PEGGuinea pig with spinal cord transectionRepairing cell membrane, and reducing oxidative stress[148]
PEGDSPESD rats with compression injury of spinal cordReducing material concentration and improving dysfunction after injury[149]
PLABone marrow stromal cellsSD rats with spinal cord transectionPromoting nerve regeneration as well as restoration of conduction, and providing a better microenvironment[150]
PLADHASD rats with spinal cord hemisectionPromoting axon regeneration with strong mechanical properties[151]
PLAAligned microfiber-based graftsSD rats with spinal cord transectionReducing cyst volume in SCI[152]
PLGADC-CholSD rats with spinal cord injuryPromoting the regeneration of blood vessels and tissues and improving exercise capacity[153]
PLGAHOMSCSD rats with spinal cord transectionPromoting endogenous repair, thereby restoring exercise capacity[154]
PLGADPSCsRats with spinal cord transectionEnhancing the regeneration of blood vessels and axons[155]
PLGAAntiNgRSD rats with spinal cord hemisectionInhibiting inflammation and promoting angiogenesis[156]
PCLHEnSCs and hSCSD rats with spinal cord hemisectionLimiting secondary reactions and restoring motor function[157]
PCLPDOSD rats with spinal cord hemisectionPromoting axonal regeneration and inhibiting the activity of astrocytes[158]
PCLFischer 344 rats with spinal cord transectionPromoting axonal growth as well as reducing scar tissue[159]
PVASD rats with spinal cord cervical contusionReducing inflammation and reducing the number of cell death—promoting proteins[160]
PVAMoS 2/GOC57BL/6 N mice with spinal cord hemisectionInhibiting inflammatory and the activation of glial cells at the site of injury[161]
PHBSD rats with spinal cord hemisectionPromoting differentiation of astrocytes but inhibits their activation[162]
PHBSD rats with spinal cord contusionInhibiting inflammatory bodies, reducing glial scar formation and promoting axonal growth[163]
PHBSD Rats with spinal cord hemi-contusionReducing neuroinflammatory reaction and improving exercise ability[164]
PSAMPSD Rats with spinal cord transectionInhibiting acute tissue inflammation and apoptosis, and promoting axon regeneration[165]
PSAHNK-1C57BL/6J mice with spinal cord compression injuryPromoting the formation of axonal myelin sheath and improving the recovery of function[166]
PSA5-NOTC57BL/6J mice with spinal cord compression injuryPromoting survival and neurite length of cortical neurons, and improving motor function[167]
pHEMAVarious substrates and nutritionalSprague–Dawley rats with spinal cord transectionIncreasing the regeneration of the damaged spinal cord and improving motor ability[168]
pHEMAWister rats with spinal cord hemisection or transectionPromoting regeneration of axons as well as blood vessels[169]
pHEMASprague–Dawley rats with spinal cord transectionPromoting axonal growth and reducing scar formation[170]
–: There is no implanted substance
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

Feng, C.; Deng, L.; Yong, Y.-Y.; Wu, J.-M.; Qin, D.-L.; Yu, L.; Zhou, X.-G.; Wu, A.-G. The Application of Biomaterials in Spinal Cord Injury. Int. J. Mol. Sci. 2023, 24, 816. https://doi.org/10.3390/ijms24010816

AMA Style

Feng C, Deng L, Yong Y-Y, Wu J-M, Qin D-L, Yu L, Zhou X-G, Wu A-G. The Application of Biomaterials in Spinal Cord Injury. International Journal of Molecular Sciences. 2023; 24(1):816. https://doi.org/10.3390/ijms24010816

Chicago/Turabian Style

Feng, Chi, Lan Deng, Yuan-Yuan Yong, Jian-Ming Wu, Da-Lian Qin, Lu Yu, Xiao-Gang Zhou, and An-Guo Wu. 2023. "The Application of Biomaterials in Spinal Cord Injury" International Journal of Molecular Sciences 24, no. 1: 816. https://doi.org/10.3390/ijms24010816

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