Next Article in Journal
Sialic Acid Ameliorates Cognitive Deficits by Reducing Amyloid Deposition, Nerve Fiber Production, and Neuronal Apoptosis in a Mice Model of Alzheimer’s Disease
Next Article in Special Issue
Transmembrane 29 (Tmem29), a Newly Identified Molecule Showed Downregulation in Hypoxic-Ischemic Brain Damage
Previous Article in Journal / Special Issue
GSK3β Activity in Reward Circuit Functioning and Addiction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Blood-Spinal Cord Barrier: Its Role in Spinal Disorders and Emerging Therapeutic Strategies

1
Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia
2
Spine Service, St. George Hospital, Kogarah, NSW 2217, Australia
3
Centre for Inflammation, Faculty of Science, Centenary Institute, School of Life Sciences, University of Technology Sydney, Sydney, NSW 2050, Australia
*
Author to whom correspondence should be addressed.
NeuroSci 2022, 3(1), 1-27; https://doi.org/10.3390/neurosci3010001
Submission received: 18 November 2021 / Revised: 9 December 2021 / Accepted: 14 December 2021 / Published: 21 December 2021
(This article belongs to the Special Issue Feature Papers in Neurosci 2021)

Abstract

:
The blood-spinal cord barrier (BSCB) has been long thought of as a functional equivalent to the blood-brain barrier (BBB), restricting blood flow into the spinal cord. The spinal cord is supported by various disc tissues that provide agility and has different local immune responses compared to the brain. Though physiologically, structural components of the BSCB and BBB share many similarities, the clinical landscape significantly differs. Thus, it is crucial to understand the composition of BSCB and also to establish the cause–effect relationship with aberrations and spinal cord dysfunctions. Here, we provide a descriptive analysis of the anatomy, current techniques to assess the impairment of BSCB, associated risk factors and impact of spinal disorders such as spinal cord injury (SCI), amyotrophic lateral sclerosis (ALS), peripheral nerve injury (PNI), ischemia reperfusion injury (IRI), degenerative cervical myelopathy (DCM), multiple sclerosis (MS), spinal cavernous malformations (SCM) and cancer on BSCB dysfunction. Along with diagnostic and mechanistic analyses, we also provide an up-to-date account of available therapeutic options for BSCB repair. We emphasize the need to address BSCB as an individual entity and direct future research towards it.

1. Introduction

Blood vessels are essential for delivering oxygen and nutrients throughout the body. In the vascular tree, the controlled communication that occurs between blood vessels and components of central nervous system (CNS) is unique. Physiologically, there are three specialised interfaces in the human body that selectively permit entry of nutrients, ions, lipids and small molecules from the blood stream to either the brain (blood-brain barrier; BBB), cerebral spinal fluid (blood-cerebral spinal fluid barrier; BCSFB) or spinal cord (blood-spinal cord barrier; BSCB). Of these the BBB is the most studied and its dysfunction is associated with neurological disorders such as multiple sclerosis (MS), Alzheimer’s disease. and Parkinson’s disease [1]. Recent evidence suggests that BBB dysfunction is an underlying mechanism associated with age-related neuronal deterioration [2]. As a result of improved understanding of the morphology and the consequences of dysfunction of the BBB, various translational drugs and models have been realised.
Drugs such as Natalizumab, a humanized monoclonal antibody acting on the VLA-4/VCAM-1 axis, modulates BBB leakage and inhibits the entry of T cells into the CNS in relapsing MS patients [3]. In Alzheimer’s disease, “Trojan horse” strategies have been used, wherein a two-sided antibody crosses the BBB and degrades β-secretase (a precursor to amyloid β protein) while remaining bound to transferrin receptor with one arm [4]. Clinical trials using strategies such as ultrasound waves to deliver drugs across the BBB are being tested for glioblastoma patients [5]. More recently, in vitro “organoids” representing BBB dysfunction have been developed as a potential platform for drug testing and development of therapeutics [6].
Although the role of BBB is well studied in brain disorders, its functional equivalent, the BSCB, lacks in-depth investigation regarding its role in spinal cord and neurological disorders as it is still considered as simply an extension of the BBB. Here, it is notable that although the constituents of both the BBB and BSCB are similar, yet functional differences in their role in diseases of the spinal cord differ [7]. Compared to the brain, the spinal cord is a far more agile organ that is surrounded by supporting diverse tissues and extracellular matrices. In the event of dysfunction, immune responses generated from the spinal cord clinically differ from those in the brain and require a suitable intervention. Dysfunction of the BSCB is associated with various traumatic and non-traumatic injuries, aging and cancer and should be considered as a separate entity for research and clinical purposes. The vascular structure of the BSCB, imaging techniques and role in diseases have been elegantly summarised by Bartanusz et al. [7]. However, over the last decade, further scientific revelations provide further detailed insight of the role of BSCB in various neurodegenerative diseases and warrants an update. Here, we fill the existing gap in the literature pertaining to the role the BSCB and BSBC in maintaining spinal cord health and how its dysfunction leads to different disorders.

2. Anatomy of BSCB

The structural components of BSCB are (see Figure 1):

2.1. Spinal Cord Microvascular Endothelial Cells (SCMECs)

These are the outermost cellular structures that form the first point of contact for any incoming molecule and restrict or allow entry of molecules between the blood and spinal cord. SCMECs act like molecular gateways and restrict the trans-flow of molecules from the blood stream to the spinal cord [8]. Endothelial cells are sealed by tight junctions (TJs) and have limited endocytic vessels. The presence of more glycogen deposits in the spinal cord than the cerebral vasculature could be related to high metabolic activity due to increased number of mitochondria in SCMECs [7].

2.2. Basal Membrane

The basal membrane or basal lamina comprise mainly of laminin, collagens, fibronectins, proteoglycans that provide structural integrity to the abluminal surface of SCMECs. It also engulfs and separates pericytes from endothelial cells and plays a role in blocking the entry of macromolecules [9].

2.3. Pericytes

Pericytes are small vessel-walled cells which are crucial for the maintenance of the BSCB. They influence the proliferation, migration, and differentiation of endothelial cells by forming the TJs and flow of soluble factors. Furthermore, pericytes are also associated with the expression and alignment of TJ proteins and reduced uptake of circulating macromolecules thereby maintaining the integrity of the BSCB [1,10,11]. Pericytes are a heterogenous cell population and a universal marker for their identification has not been defined. The most commonly used markers are aminopeptidase-N (CD13) and platelet-derived growth factor receptor (PDGFR) [8].

2.4. Astrocytic Feet

Adjoining the basal lamina, are astrocytic foot processes that maintain the functional and structural integrity of the barrier characteristics such as polarity, permeability and even re-vascularization [9,12]. These spinal-cord-astrocytes end foot processes also impart neuroprotective mechanisms to SCMECs and also highly express aquaporin-4 and potassium channels Kir4.1 that regulate resting potassium ion conductance and fluid volume in the spinal cord [7].

2.5. TJ Proteins

TJs are diffusion barriers situated between endothelial cells. TJ proteins can be categorised into (i) claudins (~27 member proteins); (ii) TJ-associated MARVEL proteins (TAMPs; MARVEL motif, occluding, tricellulin, MarvelD3); (iii) Immunoglobulin superfamily membrane proteins (JAM-A/-B/-C, coxsackie adenovirus receptor, endothelial cell-selective adhesion molecule (ESAM)) [13]. Of these, claudin-1, claudin-5 and occludin, as well as Zonula occludens (ZO-1) primarily constitute the BSCB [8]. The relative higher permeability of the BSCB compared to the BBB has been attributed to the reduced levels of specific TJ proteins such as occludin and ZO-1 [7]. Furthermore, disruptions in the expression of these TJ proteins are associated with BSCB leakage and the onset of various neurological disorders [14]. Regulatory pathways such as GSK3β are regulators of TJ protein transcription and translation via transcription factor enhancers such as catenin or suppressors of the snail family of zinc finger transcription factors [8].

2.6. Transporters

Though further research is required to identify key transporters; a few studies have identified high expression of the ABC efflux transporter-ABCA2 in the spinal cord [15,16]. More recently, Uchida et al., compared the abundance of transporter proteins in different regions of the brain and showed expression of identical proteins within a twofold range in the BBB and BSCB whilst many other proteins had >twofold lower expression in the BSCB [17]. Their key findings suggest higher expression of receptors such as insulin receptor (INSR), low-density lipoprotein receptor-related protein 1 (LRP1) and GLUT1 at corticol BBB than BBB and BSCB white matter. Aquaporins (AQPs) are important membrane proteins that regulate water movement through the BSCB. Of these, AQP-1 and -4 are key transporters that mediate water absorption in hypo-osmotic conditions [18].
For a better understanding, the morphological and physiological differences between BBB and BSCB have been highlighted in Table 1.

3. Methods to Assess BSCB Impairment

The BSCB is relatively more permeable compared to the BBB [22]. This can be attributed to differences in the composition of TJ proteins that allows secretory factors to transverse through the interface. This phenomenon was initially evidenced by Prockop et al., wherein elegant experiments showed that the kinetic transfer of [3H]-D-mannitol and [14C]-carboxyl-inulin was higher in regions of the spinal cord compared to the brain in rabbits [19]. Further, permeability of various other cytokines such as IFN-α/γ and TNF-α was higher in spinal cord compared to brain regions [23]. In order to assess the BSCB dysfunction, various tracer dyes and imaging techniques have been used.

3.1. Dye Extravasation

Evans blue dye is one of the most extensively used markers/tracers to detect BSCB leakage owing to its strong affinity towards serum albumin minimizing false-negative staining [24]. Extravasation of Evans blue has been used as evidence of BSCB disruption in models of experimental autoimmune encephalomyelitis (EAE) and spinal cord injury (SCI) [25,26]. However, additional evidence shows that Evans blue can bind to other proteins such as globulins, α1-lipoprotein, hemopexin and transferrin raising concerns over detection of barrier leaks [27]. Furthermore, toxicity concerns of Evans blue are also a major limitation for in vivo studies. Over the years, less toxic dyes such as sodium fluorescein (Na-F) have shown greater sensitivity in predicting BBB/BSCB leakage in amyotrophic lateral sclerosis (ALS) models and superiority over fluorescent tracers like fluorescein isothiocyanate (FITC)-labelled albumin and FITC-labelled dextrans-70 [28]. As well as dyes, estimating immunoglobulins (Ig) has also been used as a measure of BSCB dysfunction in degenerative cervical myelopathy (DCM) [29] and ALS [30].

3.2. Contrast Magnetic Resonance imaging (MRI)

In vivo imaging of the spinal cord is technically challenging given its small size and related motion artifacts associated with cardiovascular and respiratory organs [31]. This is a major reason for limited studies in this field thereby impacting clinical translation. In this respect, a recent systematic review by Bakhsheshian et al. evaluated various in vivo imaging techniques used to assess different diseases. They found that MRI and intravital microscopy (IVM), are the most common techniques used to assess the BSCB in rodent models of EAE and SCI [32]. Dynamic contrast-enhanced MRI (DCE-MRI) is another sensitive, non-invasive technique that can be used to assess BSCB permeability in mouse models with peripheral nerve injury (PNI) [33].

3.3. Immunohistochemistry (IHC)

The cause and effect of BSCB leakage in SCI has been assessed with infiltration of immune cells and activation of certain pathways (e.g., shh/Gli) using techniques such as IHC [34,35]. Using IHC, a direct correlation between expression of vimentin in spinal endothelium and astrocytes and pain has been established in nerve injuries [36]. A variety of other molecules such as Amigo2, aquaporin 4, CD3, CD34, GFAP, ionized calcium-binding adapter molecule 1 (Iba1), myelin basic protein (MBP), non-phosphorylated neurofilaments (np-NF), periaxin, S100A10, CCL2 and TMEV have been evaluated using IHC in cervical and thoracic spinal cord segments in Theiler’s murine EAE [37,38]. Vascular disturbances in ALS were captured by IHC where microvascular density (MVD) and pericyte coverage (PC) were quantitatively evaluated to understand their role in BSCB impairment [35].

3.4. Electron Microscopy

Electron microscopy has been used to validate polarization of AQP4 in perivascular astrocytes in Theiler’s murine EAE. This technique could also provide insights into the irregular vasculature of astrocytes that contribute to disease severity [37]. Similarly, in ALS, the frequency of degenerated endothelium and pericytes, vacuolar changes in endothelial cytoplasm in these cells were evaluated in ALS patients [39]. More recently, Ying et al., used transmission electron microscopy (TEM) to evaluate the role of BDNF/TrkB-CREB signalling pathway in tread mill training mediated BSCB protection after SCI [40]. The impairment of BSCB has been implicated in the pathophysiology of various spinal-cord related disorders such as SCI and ischemia, ALS, PNI, DCM and MS. We discuss how BSCB impairment contributes to the pathogenesis of these disorders and diseases.

4. Spinal Cord Disorders

4.1. SCI

Activation of the matrix metalloprotease (MMPs) has been regarded as a triggering event for BSCB dysfunction. Under normal physiological conditions, MMPs play a role in processes such as tissue morphogenesis, angiogenesis, cell migration, wound healing and inflammation. However, their role also encompasses degradation of various components of the extracellular matrix (ECM) thereby permitting infiltration of cells, leukocytes, etc. to breach barriers such as BBB and BSCB [41,42]. MMP-3 and -9 are regulated via up-regulation of histone H3K27 demethylase Jmjd3 leading to loss of TJ proteins and increase in BSCB permeability [43,44]. Similarly, others such as MMP-8 and -12 have also shown alterations in the abundance of TJ protein and barrier permeability as an after effect of SCI [45,46]. The mechanical stress due to SCI can damage cellular components of BSCB such as the endothelial cells via enhanced expression of cation channels such as transient receptor potential vanilloid type 4 (TRPV4) [47]. Downregulation of USP-4 after SCI promotes microglial activation and neuronal inflammation (TNF-α and IL-1β) via NF-κB by attenuating the de-ubiquitination of TRAF629. These activated microglia/macrophages can modulate neural regeneration based on their polarization (M1/M2) [48]. This phenomenon has been observed in aldose reductase (AR) knock-out mice where smaller lesion areas were observed post-SCI due to induction of M2 response as compared to the wild-type group (where AR is upregulated). The authors have implied that AR can work as a switch to regulate microglia polarization to either M1 or M2 phenotype through cAMP Response Element-Binding Protein30. Apart from NF-κB signalling, Shh/Gli is another signalling pathway which is induced in reactive astrocytes post SCI [49]. A cumulative analysis of the mechanisms and key cells of the BSCB affected post SCI have been outlined in Table 2.
Ample evidence has been provided especially by Sharma et al., where a variety of neuroprotective agents such as neurotrophins, peptide hormones [78], antioxidants [79] and bradykinin antagonist [80] have been shown to attenuate BSCB disruption evidenced by significant reductions in extravasation of protein tracers (Evans blue, iodine or lanthanum tracers). In this respect, attenuation of the SUR1/TrpM4 (known to mediate haemorrhage) and MMP-9 expression using a variety of compounds such as hormones (ghrelin, 17β-estradiol) and others (protocatechuic acid, flufenamic acid) restores the BSCB by modulating the infiltration of neutrophils and macrophages/microglia [81,82,83,84] (Table 3). The recruitment of monocyte-derived macrophages, post SCI, is in fact facilitated by brain-ventricular choroid plexus (ChP), a compartment of the BCSF, indicating its co-ordinated actions with BSCB [85]. Studies have shown that modulation of IFN-γ/IFN-γR expression by ChP could boost recruitment of anti-inflammatory molecules to the site of injury and may be a novel treatment approach [86]. Furthermore, pathways such as STAT1-NF-κB have been modulated using acidic compounds such as valproic, salvianolic and oleanolic acids to reduce pro-inflammatory responses in SCI by restoring BSCB permeability [87,88,89]. Other chemical/protein compounds with therapeutic potential in SCI induced BSCB disruption have are listed in Table 3.
Delivery systems for drugs across the BSCB have evolved over the past few years to enhance the transfer of therapeutics specifically to the sites of injury. Nanoparticles are well-established drug delivery systems owing to their nano-size, drug encapsulation capability, sustained drug release and biocompatibility [159] and have been used extensively to transport drugs across BBB (see Teleanu et al. [160]). To this end, various phase I/II clinical trials have been initiated using diverse nanoparticle–drug conjugates and are underway to target brain tumours through the BBB [161]. In line with the potential of nanoparticles to transverse BBB, some studies have also shown similar results transport across the BSCB. Nanoparticles originating from both biological (exosomes) and synthetic (lipids) sources have improved motor functions and restored TJs to attenuate BSCB leakage [121,162,163]. Other types of nanoparticles ranging from metals (e.g., iron oxide, gadolinium, cobalt, gold) and polymers to lipids have shown great potential both as tracers and drug-delivery systems in SCI (see Zuidema et al. [164]). Delivery of anti-inflammatory drugs such as methylprednisolone via nanoparticles significantly reduced lesion volume and improved behavioural outcomes when compared to delivery not guided by nanoparticles [165]. Another drug, flavopiridol, delivered using Poly (lactic-co-glycolic acid), PLGA-nanoparticles also significantly reduced inflammatory factors (e.g., TNF-α, IL-1β, IL-6) and enhanced neuronal regeneration in a SCI mouse model [166]. Additionally, PLGA-nanoparticles encapsulated in hydrogel scaffolds (hyaluronan and methylcellulose) are non-toxic and can be used for sustained drug delivery to the injury site [167]. However, with limitations of nanomaterials such as toxicity and ambiguity in systemic clearance, it is prudent that other biological nanoparticles such as exosomes are also assessed as drug delivery carriers. Additionally, grey matter has been observed to be the most affected tissue in SCI and therefore it is wise to design modalities that target smaller, deeper vascular repair in the spinal cord [168]. However, these studies are limited to investigations over short time-intervals (30min-5hours) since advanced imaging techniques such as DCE-MRI have been used to show that disruption of the BSCB occurs for as long as 56 days post SCI [169].

4.2. ALS

ALS is a fatal neurodegenerative disease characterised by motor neuron degeneration in the brain and spinal cord, causing progressive paralysis and premature death typically within 3–5 years from diagnosis [170]. The majority of ALS cases (90–95%) are sporadic (sALS) and arise from an unknown origin, while the remaining 5–10% are genetically linked or familial ALS (fALS). Of fALS cases, 20% are associated with a missense mutation in the Cu/Zn superoxide dismutase 1 (SOD1) gene [171,172]. Other associated gene mutations include chromosome 9 open reading frame 72 (C9orf72), fused in sarcoma (FUS), angiogenin (ANG) and TAR DNA binding protein 43 (TDP-43) [173]. Despite genetic variance, both sALS and fALS share most clinical and pathological presentations. Early studies of ALS patients indicated possible BSCB impairment through elevated levels of cerebrospinal fluid (CSF) proteins [174,175,176,177] and greater CSF:serum albumin ratios [176,178,179,180]. Recent proteomic analyses support these results by revealing significant differences in CSF protein expression in ALS patients compared to healthy controls [181], reinforcing the importance of BSCB leakage in ALS. Multiple studies show associations between compromised BSCB structural components (astrocytes, neurons, ECs, pericytes) and ALS (Table 2). However, there is uncertainty as to whether such changes are causative or a consequence of disease progression.
As BSCB dysfunction likely contributes to ALS pathogenesis, several studies have explored different therapeutic approaches to repair and maintain BSCB integrity. Zhong et al. [135] initially utilised activated protein C (APC), an intrinsic plasma protease with anticoagulant properties, to restor TJ (ZO-1 and occludin) expression along with no IgG leakage or microhaemorrhages in SOD1G93A mice. These results were supported by Winkler et al. [134] who used an APC mutant, 5A-APC, to significantly improve BSCB integrity and delay the onset of motor impairment in SOD1G93A mice. 5A-APC treatment similarly improved BSCB structure and function by restoring normal levels of TJ proteins including ZO-1, occludin and claudin-5 as well as eliminating IgG and free iron deposits secondary to microhaemorrhage [134]. Following this, multiple studies (Table 3) investigated intravenously transplanted unmodified human bone marrow derived CD34+ (hBM34+) stem cells and their capacity to restore BSCB integrity in ALS mice [137,138,139]. This significantly reduced microhaemorrhages [137], astrogliosis, microgliosis, capillary permeability and re-established perivascular end-feed astrocytes, as well as maintained motor neuron survival and delayed disease progression [139]. Together, these studies demonstrating improved ALS disease outcomes mediated through BSCB restoration further support the central role BSCB dysfunction in ALS. Future studies should continue to explore therapeutic approaches that support and re-enforce key structures of the BSCB, restricting access to neurotoxic substances which facilitate the ALS disease process.

4.3. PNI

Disruption of BSCB has been thought to be one of the earliest events following PNI. Initial evidence by Echeverry et al. [64], showed induction of neuropathic pain was due to recruitment of spinal blood borne monocytes/macrophages as a result of BSCB leakage. This was evidenced by presence of variably sized tracers (Evans blue/sodium fluorescein) found until 4 weeks post injury. This impairment of BSCB resulted from the release of a chemokine, monocyte chemoattractant protein-1 (MCP-1), from damaged neurons which regulates permeability in a transient and restricted manner as well as acting as a trigger for microglial activation that initiates neuropathic pain [65]. This study also reported contrasting roles of anti-inflammatory cytokines like TGFβ-1 and IL-10, which had the ability to repair the BSCB leakage post PNI. To evaluate the time-course of BSCB permeability, Cahill et al. [33], used in vivo DCE-MRI technique to show late onset BSCB permeability, which lasted for only one day post-surgery. Further, to understand the impact of variable genetic make-up on BSCB permeability and pain hypersensitivity, they assessed PNI in 5 different mouse strains (B10, C57BL/6J, CD-1, A/J and BALB/c). Interestingly, differences in permeability of Evans blue were observed amongst the different strains wherein increased up-take was observed specifically in CD-1 and A/J mice post-PNI. However, no strain related genetic correlations were observed in PNI-induced tactile hypersensitivity. Although limited, these studies do clearly indicate BSCB leakage following PNI in mice. Anti-inflammatory cytokines TGF-β1 (rescues occludin and ZO-1) and IL-10 were able to shut down the openings of the BSCB following PNI [64].

4.4. IRI

Spinal cord ischemia reperfusion injury (IRI) or SI is an adverse repercussion of thoracic aortic surgery and is closely associated with BSCB dysfunction. To-date, only one study [182] has reported impairment of BSCB that is attributed to the bimodal phase of SI. This study was based on a previously established hypothesis showing TLR-4 as an important transmembrane protein to be associated with inflammation post-SI. To further elucidate the role of TLR-4 related pathways in BSCB leakage, the expression of effector molecules such as myeloid differentiation factor 88 (MyD88) and TIR domain-containing adaptor inducing IFN-β (TRIF) were also evaluated. This showed the dependence of TLR4/MyD88 microglia-dependent activation in the first phase of SI, whereas TLR4/TRIF activation was related to the late phase, with the involvement of both microglia and astrocytes. These results emphasize the need for developing drugs for variable phases of SI. These observations corroborate with extravasations of Evans blue dye that too showed a pattern of bimodal distribution in the early stage i.e., 6–18 h post-surgery and later stage 36–48 h, which was in concurred with clinical manifestations. This team was also the first to show the effectiveness of bone marrow stromal cells in reducing inflammation in the BSCB in a SI-rabbit model [148]. Here too, BSCB leakage was recorded using Evans blue extravasation at disease onset with decreases in the TJ protein occludin and increases in MMP-9 and TNF-α levels. However, on exogenous treatment with bone marrow stromal cells, increases in the levels of occludin and inhibition of MMP-9 and TNF-α were observed, indicating restoration of BSCB integrity. Such activities of stromal cells may be attributed to their inherent anti-inflammatory properties and reduce TNF-α expression as seen in lipopolysaccharide-induced in vitro/in vivo inflammation models [183].

4.5. MS

MS is an acquired autoimmune disorder that results from changes in motor function due to effects on the brain and spinal cord. This disease has been mostly studied using an experimental animal model, EAE, that represents the physiology of human inflammatory demyelinating diseases such as MS. The pathophysiology of MS involves multifocal demyelination and neuronal loss which is attributed to the influx of activated immune cells such as leukocytes, T cells and macrophages into the CNS due to the impairment of the BBB or BSCB [184]. Although, substantial evidence establishes BBB leakage as one of the major causes of MS, few studies have examined the effects of BSCB disruption. In this regard, claudin-11, a transmembrane protein of CNS barriers (BBB, BSCB, the arachnoid barrier (BAB), BCSF), was studied to decipher its involvement in membrane disruption. Experimentation showed that downregulation of claudin-11 in the brain and spinal cord capillaries of an MS patient and EAE mice suggesting its involvement in maintaining the integrity of the BBB and BSCB [70]. One study evaluated the utility of MRI to detect lesions and BSCB disruption using the EAE model and showed the greatest disruption, predominantly in the white matter, at the onset of the disease, which declined as the disease further progressed [185]. Using the same model, this mechanism was further validated by Aube et al. [71], wherein disintegration of BSCB was observed within a day of disease onset in concurrence with the GFP+ myeloid cell infiltration into the CNS that lasted for 4 days. A major observation was that permeability of the BSCB to small tracer molecules was due to the recruitment of neutrophils in the lumbar spinal cord. The role of neutrophils in BSCB disruption was validated by depleting neutrophils in the EAE mouse model using Anti-Ly6G treatment, which showed delayed manifestation of clinical symptoms of EAE and also decreased disease severity. Furthermore, the depletion of neutrophils also reduced BSCB permeability as evidenced by reduced tracer extravasation in these mice. Activated microglia also mediates BSCB impairment eventually leading to MS.
As a therapeutic agent, tetramethylpyrazine (TMP) has been suggested as an inhibitor of glial activation by modulating microglia polarization from the M1 to M2 phenotype through activation of STAT3/SOCS3 and inhibition of NF-κB signalling pathways. Further, TMP also restores TJ proteins and decreases expression of pro-inflammatory cytokines whilst increasing the expression of ant-inflammatory cytokines [26]. In EAE models, polarization of neutrophils in the CNS has a negative impact on BCSB integrity as also observed in BBB [186]. Further, compounds such as insulin-like growth factor (IGF-1) and erythropoietin significantly improved BSCB permeability and reduced axonal damage [182,187] (Table 3). Natalizumab, a human monoclonal antibody blocks interactions between α4 (on leukocytes) and its ligand on the BBB significantly reducing the disease impact [184,188]. However, its impact on spinal cord associated-MS has not been studied. Trials in future may be designed to understand its effect on the BSCB.

4.6. SCM

SCMs, also called cavernous angiomas and cavernomas, are relatively rare intramedullary vascular lesions found in 1.86 per 100,000 population and represents 5–12% of all spinal vascular malformations, and 3–5% of spinal cord lesions [189,190,191,192,193,194]. Pathologically, SCMs are characterised by well-circumscribed vascular malformations, which often appear on the spinal cord surface [195]. Histologically, SCMs consist of sinusoidal vascular spaces lined by a single layer of endothelial cells that are surrounded by loose connective tissue stroma, predisposing to haemorrhage [196]. Lesions in the spinal cord are of clinical interest as they are often surgically inaccessible and may lead to severe complications and even death [197]. Hence, a novel pharmacological approach is urgently needed for SCM patients. No studies have investigated the BSCB in SCM. However, in cerebral cavernous malformation, another type of cavernous malformation, exhibits incompetent and absence of blood-brain barrier (BBB) [198] and disrupted endothelium [199]. This shows that further investigations of the BSCB in SCM are warranted.

4.7. DCM

DCM is a progressive non-traumatic spinal cord disorder which results from compression in the neck and is typically diagnosed late due to non-specific and common symptoms that overlap with other neurological disorders [200]. The study of BSCB impairment in DCM is in its infancy and to-date only one study has detected this disruption in human subjects. Blume et al. [29], in a recently conducted prospective non-randomized study (n = 28) showed increased permeability of the BSCB in DCM patients through AlbuminQ expression in the intrathecal space as detected by concentrations of IgQs in CSF following a lumbar puncture. Interestingly, as opposed to other diseased models such as SCI and PNI, DCM showed a longer duration of clinical symptoms with disruptions of BSCB (6 months compared to 14 days–4 weeks). Differences in proteomic expression have also been observed in the CSF of dogs suffering from cervical spondylomyelopathy (CSM). CSM-affected dogs had enhanced expression of proteins responsible for actin regulation (vitamin D-binding protein, gelsolin), white matter damage and myelin degeneration (creatine kinase B-type), osteoarthritic changes (angiotensinogen, α-2-HS-glycoprotein) and osteoarthritis (SPARC, calsyntenin-1, complement C3) that are known to be associated with BSCB impairment. Treatment with corticosteroid decreased the levels of angiotensinogen, α-2-HS-glycoprotein and gelsolin and increased the expression of proteins associated with neuroprotection (transthyretin isoform 2, apolipoprotein E, cystatin C-like) and had anti-apoptotic effects (clusterin) [201]. DCM being an isolated spinal cord disorder may likely have a unique role of the BSBC. The spinal cord in the neck is subject to excessive motion the impact of motility on the vessel basement membranes and associated structures remains to be explored. An evolving concept around the causation of DCM is biomechanical and a putative role for the BSBC should be considered in the altered complex biomechanics of the area [202].

4.8. Cancers

Spinal cord cancer is a rare malignancy of the CNS. The majority (80–85%) of the CNS tumours occur in the brain with only 10–15% in the spinal cord. To date, a growing tumour mass has not been reported to be associated with disruption of the BSCB. On the contrary, impacts on the BSCB have been observed following anti-tumour treatment such as radiation therapy. Radiation injury occurs in the BBB where morphological changes in endothelial cells and TJ proteins has been observed both at early and late stages of radiation exposure [114,203,204]. Enhanced expression of the adhesion molecule, intercellular adhesion molecule-1 (ICAM-1) is implicated in the disruption of BBB integrity on irradiation [205]. ICAM-1, an important component of the TJs, has been observed in various injuries corresponding with BBB disruption and is inversely associated with barrier integrity [206,207]. To assess the effects of irradiation, Nordal et al. [208], examined the expression of ICAM-1 on BSCB disruption in rat spinal cords. They reported BSCB leakage of serum albumin that corresponded with increased expression of ICAM-1 on endothelial and glial cells 24 hrs after radiation injury. Although increased ICAM-1 expression is not an immediate response to irradiation, it is a downstream of other target genes such as various growth factors, cytokines and transcription factors which may be induced by radiation exposure. Hypoxia induced up-regulation of vascular endothelial growth factor in astrocytes has been observed post CNS radiation injury along with BSCB disruption as indicated by albumin extravasation in rat spinal cords [209,210].

5. Risk Factors

5.1. Aging

A recent finding by Piekarz et al. [211] shows significant loss (up to 41%) of alpha motor neurons (α-MNs) with increasing age in mice. In addition, loss of myelin, compromised neuronal viability, increases in the age-related inflammation marker soluble ICAM-1 and the apoptotic marker caspase-3 are all indicative of the degenerative impact of aging on the spinal cord. Furthermore, the authors observed greater permeability of BSCB to MMP-12 in aged mice which could be a contributing factor to increased apoptosis of α-MNs.

5.2. Obesity and Metabolic Syndrome

Though the impact of obesity and metabolic syndromes such as diabetes have not been studied on the BSCB, evidence from the BBB suggest that related inflammation can cause impairment. Alterations in BBB transporters such as P-glycoprotein, low density lipoprotein receptor-related protein 1 and insulin transporters along with up-regulation of MMPs affect TJ proteins thereby causing BBB permeability [212]. However, in the BSCB, the little evidence that exists suggests that spinal cord dysfunction such as in SCI may significantly increase the risk of type 2 diabetes [213]. Further studies are warranted to establish a cause–effect relationship between metabolic disorders and the BSCB.

5.3. Lifestyle

Alcohol consumption has detrimental effect on BBB endothelial cells (in vitro) leading to gap formation between TJs along with GRP78 chaperone upregulation and increased ROS production [214]. In addition, evidence related to decreased TJ proteins (ZO-1, VE-cadherin, occludin), low-density lipoprotein receptor-related protein-1, receptor for advanced glycation end products, major facilitator superfamily domain-containing protein-2a and AQP4 with increased ROS production in mouse models (APPswe/PS1De9 mice) have demonstrated increased BBB permeability [215]. This detrimental effect of alcohol can be extrapolated to BSCB given presence of low TJs in its structural composition.

5.4. Infection and Auto-Immunity

Bacterial infections leading to meningitis influence BBB permeability by allowing flow of molecules such as albumin, nucleotide-binding oligomerization domain 2 (NOD2) and inflammatory factors [216,217]. Borrelia burgdorferi, the causative bacteria of Lyme disease also impacts cell-to-cell junctions including TJs and adherens junctions thereby influencing BCSF integrity [218]. More recently, it was observed that the vascular barrier in the choroid plexus (ChP) shuts down in response to inflammatory bowel disease via up-regulation of the Wnt/β-catenin pathway. This closure has been associated with mental deficits in models of genetically driven closure of ChP endothelial cells, highlighting the importance of peripheral interactions with the BCSF [219,220]. Furthermore, viruses such as human immunodeficiency virus type 1 (HIV-1) can target and alter the morphology and function of pericytes that constitute the BBB [221]. More recently, SARS-CoV-2 spike protein reportedly affected endothelial cells in the BBB via initiation of pro-inflammatory responses ultimately leading to leakage of pathogens, immune cells and cytokines into CSF and the brain in COVID-19 patients [222,223,224,225]. Other viruses such as Zika virus have shown evidence of penetration into brain parenchyma via alterations of TJ protein expression and disruption of BBB permeability [226,227]. Though, associations of bacterial and viral infections with BSCB integrity have not been reported, low abundance of TJ proteins in the BSCB could further aggravate its integrity and expedite damage to CNS.

5.5. Environment

Exposure to traffic related pollution has detrimental effects on the BBB. Suwannasual et al. showed that exposure to a mixture of gasoline and diesel vehicle engine emissions (MVE) led to alterations in BBB integrity (reduced TJs, upregulated IL-6, TGF-β) via Angiotensin-AT1 signalling and inflammation [228]. Similar effects on TJs were observed with environmental pollutant exposures such as perfluorooctane sulfonate [229] and pollution derived Fe3O4 nanoparticles [230]. Severe BBB dysfunction in children has been observed on exposure to dioxins such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) [231] and fine particulate matter (PM2.5) [232].

6. Conclusions

The terms BBB and BSCB have been often used interchangeably to define the blood-CNS barrier. However, evolving studies, show that BBB and BSCB are “two-sides of the same coin”. Due to anatomical arrangements, the BSCB is often regarded as a mere extension of the BBB with similar structural components. Yet, lower numbers of pericytes and TJs in the BSCB make it more permeable than the BBB [20,21]. This phenomenon has been found in various spinal-cord related diseases such as SCI, PNI and SI along with neurological disorders such as ALS, MS and DCM. Such studies highlight that BSCB breach is as crucial a contributing factor to disease prognosis as is BBB. However, when compared to the BBB, BSCB investigations are limited and still lack evidence linking modulation of specific pathways or molecular/proteomic components.
MMP3/9/12-induced leakage is a crucial event in SCI, yet investigations pertaining to modulated pathways are lacking. In BBB models, elevated levels of MMP-9 break down TJ proteins and collagenase IV via the sonic hedgehog pathway (HH) transcription factor GLI-1 [233]. Furthermore, studies have also shown that the enzymatic activities of MMP9 are regulated by tissue inhibitors of metalloproteinase-1 (TIMP1) [234] and that the MMP9:TIMP1 ratio is a determinant of disease prognosis [235]. Other MMP-independent functions such as maintenance of barrier integrity is also maintained by TIMP1, which plays a key role in disease prognosis by regulating BBB permeability [236]. Considering that the BSCB is a functional equivalent of BBB, the postulated pathways such as HH and its downstream, targets such as TIMP1 and its effector molecules could be assessed to induce BSCB repair. Another interesting emerging aspect is the association of physical exercise with BBB/BSCB permeability. Strength and endurance training in an EAE model preserved TJ proteins in spinal cord tissue and also restricted the entry of autoreactive T cells into the CNS thereby maintaining BBB integrity [237]. Additionally, this study also showed inhibition of pro-inflammatory cytokines such as IFN-γ, IL-17 and IL-1β in the spinal cord strengthening the importance of physical exercise. Despite encouraging results, a few studies have also highlighted the negative outcomes of long-term exercise on BBB permeability [237,238]. Therefore, this aspect requires further clarification especially related to its influence on the BSCB interface and variable factors such as form and duration of exercise.
To further investigations and test potential therapeutic drugs on the BSCB, developing in vitro models mimicking this interface is crucial. In this regard, organoids with their 3-dimensional spatial arrangement can overcome the limitations of adherent 2-dimensional cell cultures and can be used to accurately predict the permeability of molecules/nanoparticles. Recently, organoids depicting the neurovascular BBB have been developed by Kumarasamy et al. [239] that express BBB-related genes, TJs, enzymes, proteins and structural components, and were observed to be more efficient in replicating cell–cell communication compared to 2D monocultures. Such findings open new avenues of study by enabling high-throughput screening of novel drugs and nanoparticles. Furthermore, bio-nanoparticles such as exosomes have shown great potential to transport drugs across the BBB [240]. Testing such drug-delivery vehicles to transport compounds across the BSCB are yet to be realised and should be tested.

7. Future Directions

The BSCB has gained substantial attention as an important component of the blood-CNS interface. With advancing technology, it is now important to understand the effect/causal relationship between BSCB damage and spinal disorders. To achieve a better understanding of BSCB pathophysiology, research on establishing iPSC-derived BSCB organoids would pave the way for small molecule drug screening and also realise the utility of repurposed drugs. A better understanding of the BSCB would also open new avenues for non-invasive therapeutics via delivery of nanoparticles to the spinal cord and also the brain. Evidence also suggests that lifestyle choices can metabolically regulate BSCB and more research should be focussed on this direction.

Author Contributions

Conceptualization, A.D. and N.C.; data curation, N.C., S.M. and A.D.; writing—original draft preparation, N.C.; writing—review and editing, N.C., S.M., J.P.C., P.M.H., A.D.D. and A.D.; supervision, A.D.D. and A.D. All authors have read and agreed to the published version of the manuscript.

Funding

P.M.H. is funded by a Fellowship and grants from the National Health and Medical Research Council (NHMRC) of Australia (1175134) and by UTS. A.D.D. receives unrestricted research grants from Nuvasive Australia & Baxter Inc, Education support from Globus Medical (PA). A.D.D. receives payments from Cartago Biotech, educational consultant payments from 3M & Nuvasive, research service payment from Kunovus Technologies & Merunova. This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Daneman, R.; Zhou, L.; Kebede, A.A.; Barres, B.A. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 2010, 468, 562–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Verheggen, I.C.M.; de Jong, J.J.A.; van Boxtel, M.P.J.; Gronenschild, E.; Palm, W.M.; Postma, A.A.; Jansen, J.F.A.; Verhey, F.R.J.; Backes, W.H. Increase in blood-brain barrier leakage in healthy, older adults. Geroscience 2020, 42, 1183–1193. [Google Scholar] [CrossRef] [PubMed]
  3. Hutchinson, M. Natalizumab: A new treatment for relapsing remitting multiple sclerosis. Ther. Clin. Risk Manag. 2007, 3, 259–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Yu, Y.J.; Atwal, J.K.; Zhang, Y.; Tong, R.K.; Wildsmith, K.R.; Tan, C.; Bien-Ly, N.; Hersom, M.; Maloney, J.A.; Meilandt, W.J.; et al. Therapeutic bispecific antibodies cross the blood-brain barrier in nonhuman primates. Sci. Transl. Med. 2014, 6, 261ra154. [Google Scholar] [CrossRef]
  5. Carpentier, A.; Canney, M.; Vignot, A.; Reina, V.; Beccaria, K.; Horodyckid, C.; Karachi, C.; Leclercq, D.; Lafon, C.; Chapelon, J.Y.; et al. Clinical trial of blood-brain barrier disruption by pulsed ultrasound. Sci. Transl. Med. 2016, 8, 343re342. [Google Scholar] [CrossRef]
  6. Nzou, G.; Wicks, R.T.; VanOstrand, N.R.; Mekky, G.A.; Seale, S.A.; El-Taibany, A.; Wicks, E.E.; Nechtman, C.M.; Marrotte, E.J.; Makani, V.S.; et al. Multicellular 3D Neurovascular Unit Model for Assessing Hypoxia and Neuroinflammation Induced Blood-Brain Barrier Dysfunction. Sci. Rep. 2020, 10, 9766. [Google Scholar] [CrossRef]
  7. Bartanusz, V.; Jezova, D.; Alajajian, B.; Digicaylioglu, M. The blood-spinal cord barrier: Morphology and clinical implications. Ann. Neurol. 2011, 70, 194–206. [Google Scholar] [CrossRef]
  8. Sauer, R.S.; Kirchner, J.; Yang, S.; Hu, L.; Leinders, M.; Sommer, C.; Brack, A.; Rittner, H.L. Blood-spinal cord barrier breakdown and pericyte deficiency in peripheral neuropathy. Ann. N. Y. Acad. Sci. 2017, 1405, 71–88. [Google Scholar] [CrossRef]
  9. Garbuzova-Davis, S.; Saporta, S.; Haller, E.; Kolomey, I.; Bennett, S.P.; Potter, H.; Sanberg, P.R. Evidence of compromised blood-spinal cord barrier in early and late symptomatic SOD1 mice modeling ALS. PLoS ONE 2007, 2, e1205. [Google Scholar] [CrossRef] [Green Version]
  10. Armulik, A.; Genove, G.; Mae, M.; Nisancioglu, M.H.; Wallgard, E.; Niaudet, C.; He, L.; Norlin, J.; Lindblom, P.; Strittmatter, K.; et al. Pericytes regulate the blood-brain barrier. Nature 2010, 468, 557–561. [Google Scholar] [CrossRef] [Green Version]
  11. Bell, R.D.; Winkler, E.A.; Sagare, A.P.; Singh, I.; LaRue, B.; Deane, R.; Zlokovic, B.V. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron 2010, 68, 409–427. [Google Scholar] [CrossRef] [Green Version]
  12. Whetstone, W.D.; Hsu, J.Y.; Eisenberg, M.; Werb, Z.; Noble-Haeusslein, L.J. Blood-spinal cord barrier after spinal cord injury: Relation to revascularization and wound healing. J. Neurosci. Res. 2003, 74, 227–239. [Google Scholar] [CrossRef] [Green Version]
  13. Bauer, H.C.; Krizbai, I.A.; Bauer, H.; Traweger, A. “You Shall Not Pass”-tight junctions of the blood brain barrier. Front. Neurosci. 2014, 8, 392. [Google Scholar] [CrossRef]
  14. Katsuno, T.; Umeda, K.; Matsui, T.; Hata, M.; Tamura, A.; Itoh, M.; Takeuchi, K.; Fujimori, T.; Nabeshima, Y.; Noda, T.; et al. Deficiency of zonula occludens-1 causes embryonic lethal phenotype associated with defected yolk sac angiogenesis and apoptosis of embryonic cells. Mol. Biol. Cell. 2008, 19, 2465–2475. [Google Scholar] [CrossRef] [Green Version]
  15. Koehn, L.M.; Noor, N.M.; Dong, Q.; Er, S.Y.; Rash, L.D.; King, G.F.; Dziegielewska, K.M.; Saunders, N.R.; Habgood, M.D. Selective inhibition of ASIC1a confers functional and morphological neuroprotection following traumatic spinal cord injury. F1000Res. 2016, 5, 1822. [Google Scholar] [CrossRef]
  16. Nishimura, M.; Naito, S. Tissue-specific mRNA expression profiles of human ATP-binding cassette and solute carrier transporter superfamilies. Drug Metab. Pharmacokinet. 2005, 20, 452–477. [Google Scholar] [CrossRef]
  17. Uchida, Y.; Yagi, Y.; Takao, M.; Tano, M.; Umetsu, M.; Hirano, S.; Usui, T.; Tachikawa, M.; Terasaki, T. Comparison of Absolute Protein Abundances of Transporters and Receptors among Blood-Brain Barriers at Different Cerebral Regions and the Blood-Spinal Cord Barrier in Humans and Rats. Mol. Pharm. 2020, 17, 2006–2020. [Google Scholar] [CrossRef]
  18. Halsey, A.M.; Conner, A.C.; Bill, R.M.; Logan, A.; Ahmed, Z. Aquaporins and Their Regulation after Spinal Cord Injury. Cells 2018, 7, 174. [Google Scholar] [CrossRef] [Green Version]
  19. Prockop, L.D.; Naidu, K.A.; Binard, J.E.; Ransohoff, J. Selective permeability of [3H]-D-mannitol and [14C]-carboxyl-inulin across the blood-brain barrier and blood-spinal cord barrier in the rabbit. J. Spinal Cord Med. 1995, 18, 221–226. [Google Scholar] [CrossRef]
  20. Ge, S.; Pachter, J.S. Isolation and culture of microvascular endothelial cells from murine spinal cord. J. Neuroimmunol. 2006, 177, 209–214. [Google Scholar] [CrossRef]
  21. Winkler, E.A.; Sengillo, J.D.; Bell, R.D.; Wang, J.; Zlokovic, B.V. Blood-spinal cord barrier pericyte reductions contribute to increased capillary permeability. J. Cereb. Blood Flow Metab. 2012, 32, 1841–1852. [Google Scholar] [CrossRef] [Green Version]
  22. Wilhelm, I.; Nyul-Toth, A.; Suciu, M.; Hermenean, A.; Krizbai, I.A. Heterogeneity of the blood-brain barrier. Tissue Barriers 2016, 4, e1143544. [Google Scholar] [CrossRef]
  23. Pan, W.; Banks, W.A.; Kastin, A.J. Permeability of the blood-brain and blood-spinal cord barriers to interferons. J. Neuroimmunol. 1997, 76, 105–111. [Google Scholar] [CrossRef]
  24. Patterson, C.E.; Rhoades, R.A.; Garcia, J.G. Evans blue dye as a marker of albumin clearance in cultured endothelial monolayer and isolated lung. J. Appl. Physiol. 1992, 72, 865–873. [Google Scholar] [CrossRef]
  25. Yue, Y.; Zhao, J.; Li, X.; Zhang, L.; Su, Y.; Fan, H. Involvement of Shh/Gli1 signaling in the permeability of blood-spinal cord barrier and locomotion recovery after spinal cord contusion. Neurosci. Lett. 2020, 728, 134947. [Google Scholar] [CrossRef]
  26. Zhang, L.; Lu, X.; Gong, L.; Cui, L.; Zhang, H.; Zhao, W.; Jiang, P.; Hou, G.; Hou, Y. Tetramethylpyrazine Protects Blood-Spinal Cord Barrier Integrity by Modulating Microglia Polarization Through Activation of STAT3/SOCS3 and Inhibition of NF-small ka, CyrillicB Signaling Pathways in Experimental Autoimmune Encephalomyelitis Mice. Cell. Mol. Neurobiol. 2021, 41, 717–731. [Google Scholar] [CrossRef]
  27. Saunders, N.R.; Dziegielewska, K.M.; Mollgard, K.; Habgood, M.D. Markers for blood-brain barrier integrity: How appropriate is Evans blue in the twenty-first century and what are the alternatives? Front. Neurosci. 2015, 9, 385. [Google Scholar] [CrossRef] [Green Version]
  28. Kumar, V.; Lee, J.D.; Coulson, E.J.; Woodruff, T.M. A validated quantitative method for the assessment of neuroprotective barrier impairment in neurodegenerative disease models. J. Neurochem. 2021, 158, 807–817. [Google Scholar] [CrossRef] [PubMed]
  29. Blume, C.; Geiger, M.F.; Brandenburg, L.O.; Muller, M.; Mainz, V.; Kalder, J.; Albanna, W.; Clusmann, H.; Mueller, C.A. Patients with degenerative cervical myelopathy have signs of blood spinal cord barrier disruption, and its magnitude correlates with myelopathy severity: A prospective comparative cohort study. Eur. Spine J. 2020, 29, 986–993. [Google Scholar] [CrossRef]
  30. Winkler, E.A.; Sengillo, J.D.; Sullivan, J.S.; Henkel, J.S.; Appel, S.H.; Zlokovic, B.V. Blood-spinal cord barrier breakdown and pericyte reductions in amyotrophic lateral sclerosis. Acta Neuropathol. 2013, 125, 111–120. [Google Scholar] [CrossRef] [Green Version]
  31. Bilgen, M.; Dogan, B.; Narayana, P.A. In vivo assessment of blood-spinal cord barrier permeability: Serial dynamic contrast enhanced MRI of spinal cord injury. Magn. Reson. Imaging 2002, 20, 337–341. [Google Scholar] [CrossRef]
  32. Bakhsheshian, J.; Strickland, B.A.; Mack, W.J.; Zlokovic, B.V. Investigating the blood-spinal cord barrier in preclinical models: A systematic review of in vivo imaging techniques. Spinal Cord 2021, 59, 596–612. [Google Scholar] [CrossRef] [PubMed]
  33. Cahill, L.S.; Laliberte, C.L.; Liu, X.J.; Bishop, J.; Nieman, B.J.; Mogil, J.S.; Sorge, R.E.; Jones, C.D.; Salter, M.W.; Henkelman, R.M. Quantifying blood-spinal cord barrier permeability after peripheral nerve injury in the living mouse. Mol. Pain 2014, 10, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hellenbrand, D.J.; Reichl, K.A.; Travis, B.J.; Filipp, M.E.; Khalil, A.S.; Pulito, D.J.; Gavigan, A.V.; Maginot, E.R.; Arnold, M.T.; Adler, A.G.; et al. Sustained interleukin-10 delivery reduces inflammation and improves motor function after spinal cord injury. J. Neuroinflamm. 2019, 16, 93. [Google Scholar] [CrossRef]
  35. Yamadera, M.; Fujimura, H.; Inoue, K.; Toyooka, K.; Mori, C.; Hirano, H.; Sakoda, S. Microvascular disturbance with decreased pericyte coverage is prominent in the ventral horn of patients with amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Frontotemporal Degener. 2015, 16, 393–401. [Google Scholar] [CrossRef]
  36. Smith, J.R.; Lee, J.; Winkelstein, B.A. Nerve Root Compression Increases Spinal Astrocytic Vimentin in Parallel With Sustained Pain and Endothelial Vimentin in Association With Spinal Vascular Reestablishment. Spine 2017, 42, 1434–1439. [Google Scholar] [CrossRef]
  37. Allnoch, L.; Baumgartner, W.; Hansmann, F. Impact of Astrocyte Depletion upon Inflammation and Demyelination in a Murine Animal Model of Multiple Sclerosis. Int. J. Mol. Sci. 2019, 20, 3922. [Google Scholar] [CrossRef] [Green Version]
  38. Shrestha, B.; Ge, S.; Pachter, J.S. Resolution of central nervous system astrocytic and endothelial sources of CCL2 gene expression during evolving neuroinflammation. Fluids Barriers CNS 2014, 11, 6. [Google Scholar] [CrossRef] [Green Version]
  39. Sasaki, S. Alterations of the blood-spinal cord barrier in sporadic amyotrophic lateral sclerosis. Neuropathology 2015, 35, 518–528. [Google Scholar] [CrossRef]
  40. Ying, X.; Xie, Q.; Yu, X.; Li, S.; Wu, Q.; Chen, X.; Yue, J.; Zhou, K.; Tu, W.; Jiang, S. Water treadmill training protects the integrity of the blood-spinal cord barrier following SCI via the BDNF/TrkB-CREB signalling pathway. Neurochem. Int. 2021, 143, 104945. [Google Scholar] [CrossRef]
  41. Cawston, T.E.; Young, D.A. Proteinases involved in matrix turnover during cartilage and bone breakdown. Cell Tissue Res. 2010, 339, 221–235. [Google Scholar] [CrossRef]
  42. Lu, P.; Takai, K.; Weaver, V.M.; Werb, Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a005058. [Google Scholar] [CrossRef]
  43. Lee, J.Y.; Choi, H.Y.; Ahn, H.J.; Ju, B.G.; Yune, T.Y. Matrix metalloproteinase-3 promotes early blood-spinal cord barrier disruption and hemorrhage and impairs long-term neurological recovery after spinal cord injury. Am. J. Pathol. 2014, 184, 2985–3000. [Google Scholar] [CrossRef]
  44. Lee, J.Y.; Na, W.H.; Choi, H.Y.; Lee, K.H.; Ju, B.G.; Yune, T.Y. Jmjd3 mediates blood-spinal cord barrier disruption after spinal cord injury by regulating MMP-3 and MMP-9 expressions. Neurobiol. Dis. 2016, 95, 66–81. [Google Scholar] [CrossRef]
  45. Kumar, H.; Jo, M.J.; Choi, H.; Muttigi, M.S.; Shon, S.; Kim, B.J.; Lee, S.H.; Han, I.B. Matrix Metalloproteinase-8 Inhibition Prevents Disruption of Blood-Spinal Cord Barrier and Attenuates Inflammation in Rat Model of Spinal Cord Injury. Mol. Neurobiol. 2018, 55, 2577–2590. [Google Scholar] [CrossRef]
  46. Wells, J.E.; Rice, T.K.; Nuttall, R.K.; Edwards, D.R.; Zekki, H.; Rivest, S.; Yong, V.W. An adverse role for matrix metalloproteinase 12 after spinal cord injury in mice. J. Neurosci. 2003, 23, 10107–10115. [Google Scholar] [CrossRef] [Green Version]
  47. Kumar, H.; Lim, C.S.; Choi, H.; Joshi, H.P.; Kim, K.T.; Kim, Y.H.; Park, C.K.; Kim, H.M.; Han, I.B. Elevated TRPV4 Levels Contribute to Endothelial Damage and Scarring in Experimental Spinal Cord Injury. J. Neurosci. 2020, 40, 1943–1955. [Google Scholar] [CrossRef]
  48. Jiang, X.; Yu, M.; Ou, Y.; Cao, Y.; Yao, Y.; Cai, P.; Zhang, F. Downregulation of USP4 Promotes Activation of Microglia and Subsequent Neuronal Inflammation in Rat Spinal Cord After Injury. Neurochem. Res. 2017, 42, 3245–3253. [Google Scholar] [CrossRef]
  49. Zhang, Q.; Bian, G.; Chen, P.; Liu, L.; Yu, C.; Liu, F.; Xue, Q.; Chung, S.K.; Song, B.; Ju, G.; et al. Aldose Reductase Regulates Microglia/Macrophages Polarization Through the cAMP Response Element-Binding Protein After Spinal Cord Injury in Mice. Mol. Neurobiol. 2016, 53, 662–676. [Google Scholar] [CrossRef] [Green Version]
  50. Pan, Y.L.; Guo, Y.; Ma, Y.; Wang, L.; Zheng, S.Y.; Liu, M.M.; Huang, G.C. Aquaporin-4 expression dynamically varies after acute spinal cord injury-induced disruption of blood spinal cord barrier in rats. Neuropathology 2019, 39, 181–186. [Google Scholar] [CrossRef]
  51. Yoshizaki, S.; Tamaru, T.; Hara, M.; Kijima, K.; Tanaka, M.; Konno, D.J.; Matsumoto, Y.; Nakashima, Y.; Okada, S. Microglial inflammation after chronic spinal cord injury is enhanced by reactive astrocytes via the fibronectin/beta1 integrin pathway. J. Neuroinflamm. 2021, 18, 12. [Google Scholar] [CrossRef]
  52. Kitchen, P.; Salman, M.M.; Halsey, A.M.; Clarke-Bland, C.; MacDonald, J.A.; Ishida, H.; Vogel, H.J.; Almutiri, S.; Logan, A.; Kreida, S.; et al. Targeting Aquaporin-4 Subcellular Localization to Treat Central Nervous System Edema. Cell 2020, 181, 784–799 e719. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, Z.; Zhang, H.; Xia, H.; Wang, B.; Zhang, R.; Zeng, Q.; Guo, L.; Shen, K.; Wang, B.; Zhong, Y.; et al. CD8 T cell-derived perforin aggravates secondary spinal cord injury through destroying the blood-spinal cord barrier. Biochem. Biophys. Res. Commun. 2019, 512, 367–372. [Google Scholar] [CrossRef]
  54. Ouali Alami, N.; Tang, L.; Wiesner, D.; Commisso, B.; Bayer, D.; Weishaupt, J.; Dupuis, L.; Wong, P.; Baumann, B.; Wirth, T.; et al. Multiplexed chemogenetics in astrocytes and motoneurons restore blood-spinal cord barrier in ALS. Life Sci. Alliance 2020, 3, e201900571. [Google Scholar] [CrossRef]
  55. Garbuzova-Davis, S.; Haller, E.; Saporta, S.; Kolomey, I.; Nicosia, S.V.; Sanberg, P.R. Ultrastructure of blood-brain barrier and blood-spinal cord barrier in SOD1 mice modeling ALS. Brain Res. 2007, 1157, 126–137. [Google Scholar] [CrossRef]
  56. Mohamed, L.A.; Markandaiah, S.S.; Bonanno, S.; Pasinelli, P.; Trotti, D. Excess glutamate secreted from astrocytes drives upregulation of P-glycoprotein in endothelial cells in amyotrophic lateral sclerosis. Exp. Neurol. 2019, 316, 27–38. [Google Scholar] [CrossRef]
  57. Chan, G.N.; Evans, R.A.; Banks, D.B.; Mesev, E.V.; Miller, D.S.; Cannon, R.E. Selective induction of P-glycoprotein at the CNS barriers during symptomatic stage of an ALS animal model. Neurosci. Lett. 2017, 639, 103–113. [Google Scholar] [CrossRef] [Green Version]
  58. Garbuzova-Davis, S.; Woods, R.L., 3rd; Louis, M.K.; Zesiewicz, T.A.; Kuzmin-Nichols, N.; Sullivan, K.L.; Miller, A.M.; Hernandez-Ontiveros, D.G.; Sanberg, P.R. Reduction of circulating endothelial cells in peripheral blood of ALS patients. PLoS ONE 2010, 5, e10614. [Google Scholar] [CrossRef]
  59. Jablonski, M.R.; Jacob, D.A.; Campos, C.; Miller, D.S.; Maragakis, N.J.; Pasinelli, P.; Trotti, D. Selective increase of two ABC drug efflux transporters at the blood-spinal cord barrier suggests induced pharmacoresistance in ALS. Neurobiol. Dis. 2012, 47, 194–200. [Google Scholar] [CrossRef] [Green Version]
  60. Nicaise, C.; Mitrecic, D.; Demetter, P.; De Decker, R.; Authelet, M.; Boom, A.; Pochet, R. Impaired blood-brain and blood-spinal cord barriers in mutant SOD1-linked ALS rat. Brain Res. 2009, 1301, 152–162. [Google Scholar] [CrossRef] [Green Version]
  61. Sasaki, S.; Iguchi, Y.; Katsuno, M.; Sobue, G. Alterations in the blood-spinal cord barrier in TDP-43 conditional knockout mice. Neurosci. Lett. 2015, 598, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Tang, J.; Kang, Y.; Zhou, Y.; Li, X.; Lan, J.; Wu, L.; Feng, X.; Peng, Y. ALS-causing SOD1 mutants regulate occludin phosphorylation/ubiquitination and endocytic trafficking via the ITCH/Eps15/Rab5 axis. Neurobiol. Dis. 2021, 153, 105315. [Google Scholar] [CrossRef]
  63. Zhong, Z.; Deane, R.; Ali, Z.; Parisi, M.; Shapovalov, Y.; O’Banion, M.K.; Stojanovic, K.; Sagare, A.; Boillee, S.; Cleveland, D.W.; et al. ALS-causing SOD1 mutants generate vascular changes prior to motor neuron degeneration. Nat. Neurosci. 2008, 11, 420–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Echeverry, S.; Shi, X.Q.; Rivest, S.; Zhang, J. Peripheral nerve injury alters blood-spinal cord barrier functional and molecular integrity through a selective inflammatory pathway. J. Neurosci. 2011, 31, 10819–10828. [Google Scholar] [CrossRef]
  65. Zhang, J.; De Koninck, Y. Spatial and temporal relationship between monocyte chemoattractant protein-1 expression and spinal glial activation following peripheral nerve injury. J. Neurochem. 2006, 97, 772–783. [Google Scholar] [CrossRef]
  66. Oklinski, M.K.; Choi, H.J.; Kwon, T.H. Peripheral nerve injury induces aquaporin-4 expression and astrocytic enlargement in spinal cord. Neuroscience 2015, 311, 138–152. [Google Scholar] [CrossRef]
  67. Tsymbalyuk, O.; Gerzanich, V.; Mumtaz, A.; Andhavarapu, S.; Ivanova, S.; Makar, T.K.; Sansur, C.A.; Keller, A.; Nakamura, Y.; Bryan, J.; et al. SUR1, newly expressed in astrocytes, mediates neuropathic pain in a mouse model of peripheral nerve injury. Mol. Pain 2021, 17, 17448069211006603. [Google Scholar] [CrossRef]
  68. Blume, C.; Geiger, M.F.; Muller, M.; Clusmann, H.; Mainz, V.; Kalder, J.; Brandenburg, L.O.; Mueller, C.A. Decreased angiogenesis as a possible pathomechanism in cervical degenerative myelopathy. Sci. Rep. 2021, 11, 2497. [Google Scholar] [CrossRef]
  69. Vidal, P.M.; Ulndreaj, A.; Tetreault, L.; Hong, J.; Fehlings, M.G. The changes in systemic monocytes in humans undergoing surgical decompression for degenerative cervical myelopathy may influence clinical neurological recovery. J. Neuroimmunol. 2019, 336, 577024. [Google Scholar] [CrossRef]
  70. Uchida, Y.; Sumiya, T.; Tachikawa, M.; Yamakawa, T.; Murata, S.; Yagi, Y.; Sato, K.; Stephan, A.; Ito, K.; Ohtsuki, S.; et al. Involvement of Claudin-11 in Disruption of Blood-Brain, -Spinal Cord, and -Arachnoid Barriers in Multiple Sclerosis. Mol. Neurobiol. 2019, 56, 2039–2056. [Google Scholar] [CrossRef]
  71. Aube, B.; Levesque, S.A.; Pare, A.; Chamma, E.; Kebir, H.; Gorina, R.; Lecuyer, M.A.; Alvarez, J.I.; De Koninck, Y.; Engelhardt, B.; et al. Neutrophils mediate blood-spinal cord barrier disruption in demyelinating neuroinflammatory diseases. J. Immunol. 2014, 193, 2438–2454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Li, X.Q.; Lv, H.W.; Tan, W.F.; Fang, B.; Wang, H.; Ma, H. Role of the TLR4 pathway in blood-spinal cord barrier dysfunction during the bimodal stage after ischemia/reperfusion injury in rats. J. Neuroinflamm. 2014, 11, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Li, X.Q.; Chen, F.S.; Tan, W.F.; Fang, B.; Zhang, Z.L.; Ma, H. Elevated microRNA-129-5p level ameliorates neuroinflammation and blood-spinal cord barrier damage after ischemia-reperfusion by inhibiting HMGB1 and the TLR3-cytokine pathway. J. Neuroinflamm. 2017, 14, 205. [Google Scholar] [CrossRef] [Green Version]
  74. Chen, F.; Li, X.; Li, Z.; Zhou, Y.; Qiang, Z.; Ma, H. The roles of chemokine (C-X-C motif) ligand 13 in spinal cord ischemia-reperfusion injury in rats. Brain Res. 2020, 1727, 146489. [Google Scholar] [CrossRef]
  75. Jia, H.; Ma, H.; Li, Z.; Chen, F.; Fang, B.; Cao, X.; Chang, Y.; Qiang, Z. Downregulation of LncRNA TUG1 Inhibited TLR4 Signaling Pathway-Mediated Inflammatory Damage After Spinal Cord Ischemia Reperfusion in Rats via Suppressing TRIL Expression. J. Neuropathol. Exp. Neurol. 2019, 78, 268–282. [Google Scholar] [CrossRef]
  76. Wang, X.; Campos, C.R.; Peart, J.C.; Smith, L.K.; Boni, J.L.; Cannon, R.E.; Miller, D.S. Nrf2 upregulates ATP binding cassette transporter expression and activity at the blood-brain and blood-spinal cord barriers. J. Neurosci. 2014, 34, 8585–8593. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, C.; Xu, K.; Wang, Y.; Mao, Y.; Huang, Y.; Liang, Y.; Liu, Y.; Hao, J.; Gu, X.; Ma, Z.; et al. Spinal cannabinoid receptor 2 activation reduces hypersensitivity associated with bone cancer pain and improves the integrity of the blood-spinal cord barrier. Reg. Anesth. Pain Med. 2020, 45, 783–791. [Google Scholar] [CrossRef]
  78. Sharma, H.S. Neuroprotective effects of neurotrophins and melanocortins in spinal cord injury: An experimental study in the rat using pharmacological and morphological approaches. Ann. N. Y. Acad. Sci. 2005, 1053, 407–421. [Google Scholar] [CrossRef]
  79. Sharma, H.S.; Sjoquist, P.O.; Mohanty, S.; Wiklund, L. Post-injury treatment with a new antioxidant compound H-290/51 attenuates spinal cord trauma-induced c-fos expression, motor dysfunction, edema formation, and cell injury in the rat. Acta Neurochir. Suppl. 2006, 96, 322–328. [Google Scholar] [CrossRef]
  80. Sharma, H.S. A bradykinin BK2 receptor antagonist HOE-140 attenuates blood-spinal cord barrier permeability following a focal trauma to the rat spinal cord. An experimental study using Evans blue, [131]I-sodium and lanthanum tracers. Acta Neurochir. Suppl. 2000, 76, 159–163. [Google Scholar] [CrossRef]
  81. Lee, J.Y.; Choi, H.Y.; Na, W.H.; Ju, B.G.; Yune, T.Y. Ghrelin inhibits BSCB disruption/hemorrhage by attenuating MMP-9 and SUR1/TrpM4 expression and activation after spinal cord injury. Biochim. Biophys. Acta 2014, 1842, 2403–2412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Lee, J.Y.; Choi, H.Y.; Na, W.H.; Ju, B.G.; Yune, T.Y. 17beta-estradiol inhibits MMP-9 and SUR1/TrpM4 expression and activation and thereby attenuates BSCB disruption/hemorrhage after spinal cord injury in male rats. Endocrinology 2015, 156, 1838–1850. [Google Scholar] [CrossRef] [PubMed]
  83. Park, C.S.; Lee, J.Y.; Choi, H.Y.; Ju, B.G.; Youn, I.; Yune, T.Y. Protocatechuic acid improves functional recovery after spinal cord injury by attenuating blood-spinal cord barrier disruption and hemorrhage in rats. Neurochem. Int. 2019, 124, 181–192. [Google Scholar] [CrossRef] [PubMed]
  84. Yao, Y.; Xu, J.; Yu, T.; Chen, Z.; Xiao, Z.; Wang, J.; Hu, Y.; Wu, Y.; Zhu, D. Flufenamic acid inhibits secondary hemorrhage and BSCB disruption after spinal cord injury. Theranostics 2018, 8, 4181–4198. [Google Scholar] [CrossRef]
  85. Shechter, R.; Miller, O.; Yovel, G.; Rosenzweig, N.; London, A.; Ruckh, J.; Kim, K.W.; Klein, E.; Kalchenko, V.; Bendel, P.; et al. Recruitment of beneficial M2 macrophages to injured spinal cord is orchestrated by remote brain choroid plexus. Immunity 2013, 38, 555–569. [Google Scholar] [CrossRef] [Green Version]
  86. Kunis, G.; Baruch, K.; Rosenzweig, N.; Kertser, A.; Miller, O.; Berkutzki, T.; Schwartz, M. IFN-gamma-dependent activation of the brain’s choroid plexus for CNS immune surveillance and repair. Brain 2013, 136, 3427–3440. [Google Scholar] [CrossRef] [Green Version]
  87. Chen, S.; Ye, J.; Chen, X.; Shi, J.; Wu, W.; Lin, W.; Lin, W.; Li, Y.; Fu, H.; Li, S. Valproic acid attenuates traumatic spinal cord injury-induced inflammation via STAT1 and NF-kappaB pathway dependent of HDAC3. J. Neuroinflamm. 2018, 15, 150. [Google Scholar] [CrossRef]
  88. Wang, J.L.; Ren, C.H.; Feng, J.; Ou, C.H.; Liu, L. Oleanolic acid inhibits mouse spinal cord injury through suppressing inflammation and apoptosis via the blockage of p38 and JNK MAPKs. Biomed. Pharmacother. 2020, 123, 109752. [Google Scholar] [CrossRef]
  89. Yu, D.S.; Wang, Y.S.; Bi, Y.L.; Guo, Z.P.; Yuan, Y.J.; Tong, S.M.; Su, R.C.; Ge, L.H.; Wang, J.; Pan, Y.L.; et al. Salvianolic acid A ameliorates the integrity of blood-spinal cord barrier via miR-101/Cul3/Nrf2/HO-1 signaling pathway. Brain Res. 2017, 1657, 279–287. [Google Scholar] [CrossRef] [Green Version]
  90. Sahib, S.; Niu, F.; Sharma, A.; Feng, L.; Tian, Z.R.; Muresanu, D.F.; Nozari, A.; Sharma, H.S. Potentiation of spinal cord conduction and neuroprotection following nanodelivery of DL-3-n-butylphthalide in titanium implanted nanomaterial in a focal spinal cord injury induced functional outcome, blood-spinal cord barrier breakdown and edema formation. Int. Rev. Neurobiol. 2019, 146, 153–188. [Google Scholar] [CrossRef]
  91. Sharma, H.S.; Feng, L.; Muresanu, D.F.; Castellani, R.J.; Sharma, A. Neuroprotective effects of a potent bradykinin B2 receptor antagonist HOE-140 on microvascular permeability, blood flow disturbances, edema formation, cell injury and nitric oxide synthase upregulation following trauma to the spinal cord. Int. Rev. Neurobiol. 2019, 146, 103–152. [Google Scholar] [CrossRef]
  92. Zhou, X.; Yang, Y.; Wu, L.; Wang, Y.; Du, C.; Li, C.; Wang, Z.; Wang, Y. Brilliant Blue G Inhibits Inflammasome Activation and Reduces Disruption of Blood-Spinal Cord Barrier Induced by Spinal Cord Injury in Rats. Med. Sci. Monit. 2019, 25, 6359–6366. [Google Scholar] [CrossRef]
  93. Chio, J.C.T.; Wang, J.; Badner, A.; Hong, J.; Surendran, V.; Fehlings, M.G. The effects of human immunoglobulin G on enhancing tissue protection and neurobehavioral recovery after traumatic cervical spinal cord injury are mediated through the neurovascular unit. J. Neuroinflamm. 2019, 16, 141. [Google Scholar] [CrossRef]
  94. Chio, J.C.T.; Wang, J.; Surendran, V.; Li, L.; Zavvarian, M.M.; Pieczonka, K.; Fehlings, M.G. Delayed administration of high dose human immunoglobulin G enhances recovery after traumatic cervical spinal cord injury by modulation of neuroinflammation and protection of the blood spinal cord barrier. Neurobiol. Dis. 2021, 148, 105187. [Google Scholar] [CrossRef]
  95. Lin, Y.; Vreman, H.J.; Wong, R.J.; Tjoa, T.; Yamauchi, T.; Noble-Haeusslein, L.J. Heme oxygenase-1 stabilizes the blood-spinal cord barrier and limits oxidative stress and white matter damage in the acutely injured murine spinal cord. J. Cereb. Blood Flow Metab. 2007, 27, 1010–1021. [Google Scholar] [CrossRef] [Green Version]
  96. Wang, H.; Wu, Y.; Han, W.; Li, J.; Xu, K.; Li, Z.; Wang, Q.; Xu, K.; Liu, Y.; Xie, L.; et al. Hydrogen Sulfide Ameliorates Blood-Spinal Cord Barrier Disruption and Improves Functional Recovery by Inhibiting Endoplasmic Reticulum Stress-Dependent Autophagy. Front. Pharmacol. 2018, 9, 858. [Google Scholar] [CrossRef]
  97. Park, C.S.; Lee, J.Y.; Choi, H.Y.; Lee, K.; Heo, Y.; Ju, B.G.; Choo, H.P.; Yune, T.Y. Gallic acid attenuates blood-spinal cord barrier disruption by inhibiting Jmjd3 expression and activation after spinal cord injury. Neurobiol. Dis. 2020, 145, 105077. [Google Scholar] [CrossRef]
  98. Zheng, B.; Zhou, Y.; Zhang, H.; Yang, G.; Hong, Z.; Han, D.; Wang, Q.; He, Z.; Liu, Y.; Wu, F.; et al. Dl-3-n-butylphthalide prevents the disruption of blood-spinal cord barrier via inhibiting endoplasmic reticulum stress following spinal cord injury. Int. J. Biol. Sci. 2017, 13, 1520–1531. [Google Scholar] [CrossRef]
  99. He, Z.; Zhou, Y.; Wang, Q.; Li, J.; Zheng, Z.; Chen, J.; Zhang, H.; Wang, Z.; Xu, H.; Xiao, J. Inhibiting endoplasmic reticulum stress by lithium chloride contributes to the integrity of blood-spinal cord barrier and functional recovery after spinal cord injury. Am. J. Transl. Res. 2017, 9, 1012–1024. [Google Scholar]
  100. Tong, M.; He, Z.; Lin, X.; Zhou, Y.; Wang, Q.; Zheng, Z.; Chen, J.; Xu, H.; Tian, N. Lithium chloride contributes to blood-spinal cord barrier integrity and functional recovery from spinal cord injury by stimulating autophagic flux. Biochem. Biophys. Res. Commun. 2018, 495, 2525–2531. [Google Scholar] [CrossRef]
  101. Zhang, Q.; Wang, J.; Gu, Z.; Zhang, Q.; Zheng, H. Effect of lycopene on the blood-spinal cord barrier after spinal cord injury in mice. Biosci. Trends 2016, 10, 288–293. [Google Scholar] [CrossRef] [Green Version]
  102. He, Z.; Zou, S.; Yin, J.; Gao, Z.; Liu, Y.; Wu, Y.; He, H.; Zhou, Y.; Wang, Q.; Li, J.; et al. Inhibition of Endoplasmic Reticulum Stress Preserves the Integrity of Blood-Spinal Cord Barrier in Diabetic Rats Subjected to Spinal Cord Injury. Sci. Rep. 2017, 7, 7661. [Google Scholar] [CrossRef] [Green Version]
  103. Zhou, Y.; Ye, L.; Zheng, B.; Zhu, S.; Shi, H.; Zhang, H.; Wang, Z.; Wei, X.; Chen, D.; Li, X.; et al. Phenylbutyrate prevents disruption of blood-spinal cord barrier by inhibiting endoplasmic reticulum stress after spinal cord injury. Am. J. Transl. Res. 2016, 8, 1864–1875. [Google Scholar]
  104. Sanchez-Ventura, J.; Amo-Aparicio, J.; Navarro, X.; Penas, C. BET protein inhibition regulates cytokine production and promotes neuroprotection after spinal cord injury. J. Neuroinflamm. 2019, 16, 124. [Google Scholar] [CrossRef]
  105. Miranpuri, G.S.; Nguyen, J.; Moreno, N.; Yutuc, N.A.; Kim, J.; Buttar, S.; Brown, G.R.; Sauer, S.E.; Singh, C.K.; Kumar, S.; et al. Folic Acid Modulates Matrix Metalloproteinase-9 Expression Following Spinal Cord Injury. Ann. Neurosci. 2019, 26, 60–65. [Google Scholar] [CrossRef]
  106. Fan, Z.K.; Lv, G.; Wang, Y.F.; Li, G.; Yu, D.S.; Wang, Y.S.; Zhang, Y.Q.; Mei, X.F.; Cao, Y. The protective effect of salvianolic acid B on blood-spinal cord barrier after compression spinal cord injury in rats. J. Mol. Neurosci. 2013, 51, 986–993. [Google Scholar] [CrossRef]
  107. Lee, J.Y.; Choi, H.Y.; Yune, T.Y. Fluoxetine and vitamin C synergistically inhibits blood-spinal cord barrier disruption and improves functional recovery after spinal cord injury. Neuropharmacology 2016, 109, 78–87. [Google Scholar] [CrossRef]
  108. Lee, J.Y.; Kim, H.S.; Choi, H.Y.; Oh, T.H.; Yune, T.Y. Fluoxetine inhibits matrix metalloprotease activation and prevents disruption of blood-spinal cord barrier after spinal cord injury. Brain 2012, 135, 2375–2389. [Google Scholar] [CrossRef] [Green Version]
  109. Gao, K.; Shen, Z.; Yuan, Y.; Han, D.; Song, C.; Guo, Y.; Mei, X. Simvastatin inhibits neural cell apoptosis and promotes locomotor recovery via activation of Wnt/beta-catenin signaling pathway after spinal cord injury. J. Neurochem. 2016, 138, 139–149. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, K.W.; Liang, C.L.; Yeh, L.R.; Liu, K.Y.; Chen, C.C.; Chen, J.S.; Chen, H.J.; Wang, H.K. Simvastatin-Ezetimibe enhances growth factor expression and attenuates neuron loss in the hippocampus in a model of intracerebral hemorrhage. Fundam. Clin. Pharmacol. 2020, 35, 634–644. [Google Scholar] [CrossRef] [PubMed]
  111. Zhou, Y.; Zhang, H.; Zheng, B.; Ye, L.; Zhu, S.; Johnson, N.R.; Wang, Z.; Wei, X.; Chen, D.; Cao, G.; et al. Retinoic Acid Induced-Autophagic Flux Inhibits ER-Stress Dependent Apoptosis and Prevents Disruption of Blood-Spinal Cord Barrier after Spinal Cord Injury. Int. J. Biol. Sci. 2016, 12, 87–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Ozturk, A.M.; Sozbilen, M.C.; Sevgili, E.; Dagci, T.; Ozyalcin, H.; Armagan, G. Epidermal growth factor regulates apoptosis and oxidative stress in a rat model of spinal cord injury. Injury 2018, 49, 1038–1045. [Google Scholar] [CrossRef] [PubMed]
  113. Li, J.; Wang, Q.; Cai, H.; He, Z.; Wang, H.; Chen, J.; Zheng, Z.; Yin, J.; Liao, Z.; Xu, H.; et al. FGF1 improves functional recovery through inducing PRDX1 to regulate autophagy and anti-ROS after spinal cord injury. J. Cell Mol. Med. 2018, 22, 2727–2738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Pena, L.A.; Fuks, Z.; Kolesnick, R.N. Radiation-induced apoptosis of endothelial cells in the murine central nervous system: Protection by fibroblast growth factor and sphingomyelinase deficiency. Cancer Res. 2000, 60, 321–327. [Google Scholar] [PubMed]
  115. Cabrera-Aldana, E.E.; Ruelas, F.; Aranda, C.; Rincon-Heredia, R.; Martinez-Cruz, A.; Reyes-Sanchez, A.; Guizar-Sahagun, G.; Tovar, Y.R.L.B. Methylprednisolone Administration Following Spinal Cord Injury Reduces Aquaporin 4 Expression and Exacerbates Edema. Mediators Inflamm. 2017, 2017, 4792932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Fan, Z.K.; Wang, Y.F.; Cao, Y.; Zhang, M.C.; Zhang, Z.; Lv, G.; Lu, W.; Zhang, Y.Q. The effect of aminoguanidine on compression spinal cord injury in rats. Brain Res. 2010, 1342, 1–10. [Google Scholar] [CrossRef]
  117. Wang, C.; Wang, P.; Zeng, W.; Li, W. Tetramethylpyrazine improves the recovery of spinal cord injury via Akt/Nrf2/HO-1 pathway. Bioorg. Med. Chem. Lett. 2016, 26, 1287–1291. [Google Scholar] [CrossRef]
  118. Yu, D.S.; Cao, Y.; Mei, X.F.; Wang, Y.F.; Fan, Z.K.; Wang, Y.S.; Lv, G. Curcumin improves the integrity of blood-spinal cord barrier after compressive spinal cord injury in rats. J. Neurol. Sci. 2014, 346, 51–59. [Google Scholar] [CrossRef]
  119. Ma, L.; Mu, Y.; Zhang, Z.; Sun, Q. Eugenol promotes functional recovery and alleviates inflammation, oxidative stress, and neural apoptosis in a rat model of spinal cord injury. Restor. Neurol. Neurosci. 2018, 36, 659–668. [Google Scholar] [CrossRef]
  120. Lee, J.Y.; Choi, H.Y.; Park, C.S.; Ju, B.G.; Yune, T.Y. Mithramycin A Improves Functional Recovery by Inhibiting BSCB Disruption and Hemorrhage after Spinal Cord Injury. J. Neurotrauma 2018, 35, 508–520. [Google Scholar] [CrossRef]
  121. Joshi, H.P.; Kumar, H.; Choi, U.Y.; Lim, Y.C.; Choi, H.; Kim, J.; Kyung, J.W.; Sohn, S.; Kim, K.T.; Kim, J.K.; et al. CORM-2-Solid Lipid Nanoparticles Maintain Integrity of Blood-Spinal Cord Barrier After Spinal Cord Injury in Rats. Mol. Neurobiol. 2020, 57, 2671–2689. [Google Scholar] [CrossRef] [PubMed]
  122. Zheng, G.; Zheng, F.; Luo, Z.; Ma, H.; Zheng, D.; Xiang, G.; Xu, C.; Zhou, Y.; Wu, Y.; Tian, N.; et al. CO-Releasing Molecule (CORM)-3 Ameliorates Spinal Cord-Blood Barrier Disruption Following Injury to the Spinal Cord. Front. Pharmacol. 2020, 11, 761. [Google Scholar] [CrossRef] [PubMed]
  123. Lu, Y.; Zhou, Y.; Zhang, R.; Wen, L.; Wu, K.; Li, Y.; Yao, Y.; Duan, R.; Jia, Y. Bone Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Recovery Following Spinal Cord Injury via Improvement of the Integrity of the Blood-Spinal Cord Barrier. Front. Neurosci. 2019, 13, 209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Gao, S.J.; Liu, Y.; Wang, H.J.; Ban, D.X.; Cheng, S.Z.; Ning, G.Z.; Wang, L.L.; Chang, J.; Feng, S.Q. New approach to treating spinal cord injury using PEG-TAT-modified, cyclosporine-A-loaded PLGA/polymeric liposomes. J. Drug Target. 2017, 25, 75–82. [Google Scholar] [CrossRef]
  125. Gao, Y.; Vijayaraghavalu, S.; Stees, M.; Kwon, B.K.; Labhasetwar, V. Evaluating accessibility of intravenously administered nanoparticles at the lesion site in rat and pig contusion models of spinal cord injury. J. Control. Release 2019, 302, 160–168. [Google Scholar] [CrossRef]
  126. Liu, Y.; Wang, C.Y.; Kong, X.H.; Wang, H.J.; Chang, J.; Zhang, D.P.; Ban, D.X.; Feng, S.Q. Novel multifunctional polyethylene glycol-transactivating-transduction protein-modified liposomes cross the blood-spinal cord barrier after spinal cord injury. J. Drug Target. 2010, 18, 420–429. [Google Scholar] [CrossRef]
  127. Zhang, D.; Wang, Q.; Wang, S.; Huang, Y.; Tian, N.; Wu, Y.; Wu, Y.; Zhou, Y.; Xu, H.; Zhang, X. Astragoloside IV Loaded Polycaprolactone Membrane Repairs Blood Spinal Cord Barrier and Recovers Spinal Cord Function in Traumatic Spinal Cord Injury. J. Biomed. Nanotechnol. 2019, 15, 799–812. [Google Scholar] [CrossRef]
  128. Matsushita, T.; Lankford, K.L.; Arroyo, E.J.; Sasaki, M.; Neyazi, M.; Radtke, C.; Kocsis, J.D. Diffuse and persistent blood-spinal cord barrier disruption after contusive spinal cord injury rapidly recovers following intravenous infusion of bone marrow mesenchymal stem cells. Exp. Neurol. 2015, 267, 152–164. [Google Scholar] [CrossRef]
  129. Kang, C.E.; Baumann, M.D.; Tator, C.H.; Shoichet, M.S. Localized and sustained delivery of fibroblast growth factor-2 from a nanoparticle-hydrogel composite for treatment of spinal cord injury. Cells Tissues Organs 2013, 197, 55–63. [Google Scholar] [CrossRef]
  130. Wang, J.; Nie, Z.; Zhao, H.; Gao, K.; Cao, Y. MiRNA-125a-5p attenuates blood-spinal cord barrier permeability under hypoxia in vitro. Biotechnol. Lett. 2020, 42, 25–34. [Google Scholar] [CrossRef]
  131. Sun, R.; Ge, L.; Cao, Y.; Wu, W.; Wu, Y.; Zhu, H.; Li, J.; Yu, D. MiR-429 regulates blood-spinal cord barrier permeability by targeting Kruppel-like factor 6. Biochem. Biophys. Res. Commun. 2020, 525, 740–746. [Google Scholar] [CrossRef]
  132. Sun, R.; Yu, D. Inhibitory effect of miR-429 on expressions of ZO-1, Occludin, and Claudin-5 proteins to improve the permeability of blood spinal cord barrier in vitro. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2020, 34, 1163–1169. [Google Scholar] [CrossRef]
  133. Chang, S.; Bi, Y.; Meng, X.; Qu, L.; Cao, Y. Adenovirus-delivered GFP-HO-1C[INCREMENT]23 attenuates blood-spinal cord barrier permeability after rat spinal cord contusion. Neuroreport 2018, 29, 402–407. [Google Scholar] [CrossRef]
  134. Winkler, E.A.; Sengillo, J.D.; Sagare, A.P.; Zhao, Z.; Ma, Q.; Zuniga, E.; Wang, Y.; Zhong, Z.; Sullivan, J.S.; Griffin, J.H.; et al. Blood-spinal cord barrier disruption contributes to early motor-neuron degeneration in ALS-model mice. Proc. Natl. Acad. Sci. USA 2014, 111, E1035–E1042. [Google Scholar] [CrossRef] [Green Version]
  135. Zhong, Z.; Ilieva, H.; Hallagan, L.; Bell, R.; Singh, I.; Paquette, N.; Thiyagarajan, M.; Deane, R.; Fernandez, J.A.; Lane, S.; et al. Activated protein C therapy slows ALS-like disease in mice by transcriptionally inhibiting SOD1 in motor neurons and microglia cells. J. Clin. Investig. 2009, 119, 3437–3449. [Google Scholar] [CrossRef] [Green Version]
  136. Rabinovich-Nikitin, I.; Ezra, A.; Barbiro, B.; Rabinovich-Toidman, P.; Solomon, B. Chronic administration of AMD3100 increases survival and alleviates pathology in SOD1(G93A) mice model of ALS. J. Neuroinflamm. 2016, 13, 123. [Google Scholar] [CrossRef] [Green Version]
  137. Eve, D.J.; Steiner, G.; Mahendrasah, A.; Sanberg, P.R.; Kurien, C.; Thomson, A.; Borlongan, C.V.; Garbuzova-Davis, S. Reduction of microhemorrhages in the spinal cord of symptomatic ALS mice after intravenous human bone marrow stem cell transplantation accompanies repair of the blood-spinal cord barrier. Oncotarget 2018, 9, 10621–10634. [Google Scholar] [CrossRef] [Green Version]
  138. Garbuzova-Davis, S.; Haller, E.; Navarro, S.; Besong, T.E.; Boccio, K.J.; Hailu, S.; Khatib, M.; Sanberg, P.R.; Appel, S.H.; Borlongan, C.V. Transplantation of human bone marrow stem cells into symptomatic ALS mice enhances structural and functional blood-spinal cord barrier repair. Exp. Neurol. 2018, 310, 33–47. [Google Scholar] [CrossRef]
  139. Garbuzova-Davis, S.; Kurien, C.; Thomson, A.; Falco, D.; Ahmad, S.; Staffetti, J.; Steiner, G.; Abraham, S.; James, G.; Mahendrasah, A.; et al. Endothelial and Astrocytic Support by Human Bone Marrow Stem Cell Grafts into Symptomatic ALS Mice towards Blood-Spinal Cord Barrier Repair. Sci. Rep. 2017, 7, 884. [Google Scholar] [CrossRef]
  140. Garbuzova-Davis, S.; Kurien, C.; Haller, E.; Eve, D.J.; Navarro, S.; Steiner, G.; Mahendrasah, A.; Hailu, S.; Khatib, M.; Boccio, K.J.; et al. Human Bone Marrow Endothelial Progenitor Cell Transplantation into Symptomatic ALS Mice Delays Disease Progression and Increases Motor Neuron Survival by Repairing Blood-Spinal Cord Barrier. Sci. Rep. 2019, 9, 5280. [Google Scholar] [CrossRef] [Green Version]
  141. Magota, H.; Sasaki, M.; Kataoka-Sasaki, Y.; Oka, S.; Ukai, R.; Kiyose, R.; Onodera, R.; Kocsis, J.D.; Honmou, O. Intravenous infusion of mesenchymal stem cells delays disease progression in the SOD1G93A transgenic amyotrophic lateral sclerosis rat model. Brain Res. 2021, 1757, 147296. [Google Scholar] [CrossRef] [PubMed]
  142. Xie, L.J.; Huang, J.X.; Yang, J.; Yuan, F.; Zhang, S.S.; Yu, Q.J.; Hu, J. Propofol protects against blood-spinal cord barrier disruption induced by ischemia/reperfusion injury. Neural Regen. Res. 2017, 12, 125–132. [Google Scholar] [CrossRef] [PubMed]
  143. Fang, B.; Li, X.Q.; Bi, B.; Tan, W.F.; Liu, G.; Zhang, Y.; Ma, H. Dexmedetomidine attenuates blood-spinal cord barrier disruption induced by spinal cord ischemia reperfusion injury in rats. Cell Physiol. Biochem. 2015, 36, 373–383. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, J.; Zhang, S.; Fan, X.; Yuan, F.; Dai, J.; Hu, J. Dexmedetomidine Preconditioning Ameliorates Inflammation and Blood-Spinal Cord Barrier Damage After Spinal Cord Ischemia-Reperfusion Injury by Down-Regulation High Mobility Group Box 1-Toll-Like Receptor 4-Nuclear Factor kappaB Signaling Pathway. Spine 2019, 44, E74–E81. [Google Scholar] [CrossRef] [PubMed]
  145. Fang, B.; Li, X.M.; Sun, X.J.; Bao, N.R.; Ren, X.Y.; Lv, H.W.; Ma, H. Ischemic preconditioning protects against spinal cord ischemia-reperfusion injury in rabbits by attenuating blood spinal cord barrier disruption. Int. J. Mol. Sci. 2013, 14, 10343–10354. [Google Scholar] [CrossRef] [Green Version]
  146. Jing, N.; Fang, B.; Wang, Z.L.; Ma, H. Remote Ischemia Preconditioning Attenuates Blood-Spinal Cord Barrier Breakdown in Rats Undergoing Spinal Cord Ischemia Reperfusion Injury: Associated with Activation and Upregulation of CB1 and CB2 Receptors. Cell Physiol. Biochem. 2017, 43, 2516–2524. [Google Scholar] [CrossRef] [Green Version]
  147. Li, X.Q.; Cao, X.Z.; Wang, J.; Fang, B.; Tan, W.F.; Ma, H. Sevoflurane preconditioning ameliorates neuronal deficits by inhibiting microglial MMP-9 expression after spinal cord ischemia/reperfusion in rats. Mol. Brain 2014, 7, 69. [Google Scholar] [CrossRef] [Green Version]
  148. Fang, B.; Wang, H.; Sun, X.J.; Li, X.Q.; Ai, C.Y.; Tan, W.F.; White, P.F.; Ma, H. Intrathecal transplantation of bone marrow stromal cells attenuates blood-spinal cord barrier disruption induced by spinal cord ischemia-reperfusion injury in rabbits. J. Vasc. Surg. 2013, 58, 1043–1052. [Google Scholar] [CrossRef] [Green Version]
  149. Yasuda, N.; Sasaki, M.; Kataoka-Sasaki, Y.; Nagahama, H.; Kocsis, J.D.; Kawaharada, N.; Honmou, O. Intravenous delivery of mesenchymal stem cells protects both white and gray matter in spinal cord ischemia. Brain Res. 2020, 1747, 147040. [Google Scholar] [CrossRef]
  150. Wang, D.; Chen, F.; Fang, B.; Zhang, Z.; Dong, Y.; Tong, X.; Ma, H. MiR-128-3p Alleviates Spinal Cord Ischemia/Reperfusion Injury Associated Neuroinflammation and Cellular Apoptosis via SP1 Suppression in Rat. Front. Neurosci. 2020, 14, 609613. [Google Scholar] [CrossRef]
  151. Li, X.Q.; Fang, B.; Tan, W.F.; Wang, Z.L.; Sun, X.J.; Zhang, Z.L.; Ma, H. miR-320a affects spinal cord edema through negatively regulating aquaporin-1 of blood-spinal cord barrier during bimodal stage after ischemia reperfusion injury in rats. BMC Neurosci. 2016, 17, 10. [Google Scholar] [CrossRef] [Green Version]
  152. Li, X.Q.; Lv, H.W.; Wang, Z.L.; Tan, W.F.; Fang, B.; Ma, H. MiR-27a ameliorates inflammatory damage to the blood-spinal cord barrier after spinal cord ischemia: Reperfusion injury in rats by downregulating TICAM-2 of the TLR4 signaling pathway. J. Neuroinflamm. 2015, 12, 25. [Google Scholar] [CrossRef] [Green Version]
  153. De Oliveira, L.R.C.; Mimura, L.A.N.; Fraga-Silva, T.F.C.; Ishikawa, L.L.W.; Fernandes, A.A.H.; Zorzella-Pezavento, S.F.G.; Sartori, A. Calcitriol Prevents Neuroinflammation and Reduces Blood-Brain Barrier Disruption and Local Macrophage/Microglia Activation. Front. Pharmacol. 2020, 11, 161. [Google Scholar] [CrossRef]
  154. Lu, K.; Liu, L.; Xu, X.; Zhao, F.; Deng, J.; Tang, X.; Wang, X.; Zhao, B.Q.; Zhang, X.; Zhao, Y. ADAMTS13 ameliorates inflammatory responses in experimental autoimmune encephalomyelitis. J. Neuroinflamm. 2020, 17, 67. [Google Scholar] [CrossRef] [Green Version]
  155. Mondal, S.; Dasarathi, S.; Pahan, K. Glyceryl Tribenzoate: A Flavoring Ingredient, Inhibits the Adoptive Transfer of Experimental Allergic Encephalomyelitis via TGF-beta: Implications for Multiple Sclerosis Therapy. J. Clin. Cell Immunol. 2017, 8, 488. [Google Scholar] [CrossRef] [Green Version]
  156. Mondal, S.; Pahan, K. Cinnamon ameliorates experimental allergic encephalomyelitis in mice via regulatory T cells: Implications for multiple sclerosis therapy. PLoS ONE 2015, 10, e0116566. [Google Scholar] [CrossRef] [Green Version]
  157. Hou, Y.; Heon Ryu, C.; Jun, J.A.; Kim, S.M.; Jeong, C.H.; Jeun, S.S. Interferon beta-secreting mesenchymal stem cells combined with minocycline attenuate experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2014, 274, 20–27. [Google Scholar] [CrossRef]
  158. Shao, Y.; Sang, J.; Fu, J. On human pluripotent stem cell control: The rise of 3D bioengineering and mechanobiology. Biomaterials 2015, 52, 26–43. [Google Scholar] [CrossRef] [Green Version]
  159. Fanciullino, R.; Ciccolini, J.; Milano, G. Challenges, expectations and limits for nanoparticles-based therapeutics in cancer: A focus on nano-albumin-bound drugs. Crit. Rev. Oncol. Hemato.l 2013, 88, 504–513. [Google Scholar] [CrossRef]
  160. Teleanu, D.M.; Chircov, C.; Grumezescu, A.M.; Volceanov, A.; Teleanu, R.I. Blood-Brain Delivery Methods Using Nanotechnology. Pharmaceutics 2018, 10, 269. [Google Scholar] [CrossRef] [Green Version]
  161. Zottel, A.; Videtic Paska, A.; Jovcevska, I. Nanotechnology Meets Oncology: Nanomaterials in Brain Cancer Research, Diagnosis and Therapy. Materials 2019, 12, 1588. [Google Scholar] [CrossRef] [Green Version]
  162. Guo, S.; Perets, N.; Betzer, O.; Ben-Shaul, S.; Sheinin, A.; Michaelevski, I.; Popovtzer, R.; Offen, D.; Levenberg, S. Intranasal Delivery of Mesenchymal Stem Cell Derived Exosomes Loaded with Phosphatase and Tensin Homolog siRNA Repairs Complete Spinal Cord Injury. ACS Nano 2019, 13, 10015–10028. [Google Scholar] [CrossRef]
  163. Yuan, X.; Wu, Q.; Wang, P.; Jing, Y.; Yao, H.; Tang, Y.; Li, Z.; Zhang, H.; Xiu, R. Exosomes Derived From Pericytes Improve Microcirculation and Protect Blood-Spinal Cord Barrier After Spinal Cord Injury in Mice. Front. Neurosci. 2019, 13, 319. [Google Scholar] [CrossRef]
  164. Zuidema, J.M.; Gilbert, R.J.; Osterhout, D.J. Nanoparticle Technologies in the Spinal Cord. Cells Tissues Organs 2016, 202, 102–115. [Google Scholar] [CrossRef]
  165. 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] [Green Version]
  166. Ren, H.; Han, M.; Zhou, J.; Zheng, Z.F.; Lu, P.; Wang, J.J.; Wang, J.Q.; Mao, Q.J.; Gao, J.Q.; Ouyang, H.W. Repair of spinal cord injury by inhibition of astrocyte growth and inflammatory factor synthesis through local delivery of flavopiridol in PLGA nanoparticles. Biomaterials 2014, 35, 6585–6594. [Google Scholar] [CrossRef]
  167. Baumann, M.D.; Kang, C.E.; Tator, C.H.; Shoichet, M.S. Intrathecal delivery of a polymeric nanocomposite hydrogel after spinal cord injury. Biomaterials 2010, 31, 7631–7639. [Google Scholar] [CrossRef]
  168. Figley, S.A.; Khosravi, R.; Legasto, J.M.; Tseng, Y.F.; Fehlings, M.G. Characterization of vascular disruption and blood-spinal cord barrier permeability following traumatic spinal cord injury. J. Neurotrauma 2014, 31, 541–552. [Google Scholar] [CrossRef] [Green Version]
  169. Cohen, D.M.; Patel, C.B.; Ahobila-Vajjula, P.; Sundberg, L.M.; Chacko, T.; Liu, S.J.; Narayana, P.A. Blood-spinal cord barrier permeability in experimental spinal cord injury: Dynamic contrast-enhanced MRI. NMR Biomed. 2009, 22, 332–341. [Google Scholar] [CrossRef] [Green Version]
  170. Hardiman, O.; Al-Chalabi, A.; Chio, A.; Corr, E.M.; Logroscino, G.; Robberecht, W.; Shaw, P.J.; Simmons, Z.; van den Berg, L.H. Amyotrophic lateral sclerosis. Nat. Rev. Dis. Primers 2017, 3, 17085. [Google Scholar] [CrossRef]
  171. Rosen, D.R.; Siddique, T.; Patterson, D.; Figlewicz, D.A.; Sapp, P.; Hentati, A.; Donaldson, D.; Goto, J.; O’Regan, J.P.; Deng, H.X.; et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993, 362, 59–62. [Google Scholar] [CrossRef] [PubMed]
  172. Rowland, L.P.; Shneider, N.A. Amyotrophic lateral sclerosis. N. Engl. J. Med. 2001, 344, 1688–1700. [Google Scholar] [CrossRef] [PubMed]
  173. Li, H.F.; Wu, Z.Y. Genotype-phenotype correlations of amyotrophic lateral sclerosis. Transl. Neurodegener. 2016, 5, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Guiloff, R.J.; McGregor, B.; Thompson, E.; Blackwood, W.; Paul, E. Motor neurone disease with elevated cerebrospinal fluid protein. J. Neurol. Neurosurg. Psychiatry 1980, 43, 390–396. [Google Scholar] [CrossRef] [Green Version]
  175. Leonardi, A.; Abbruzzese, G.; Arata, L.; Cocito, L.; Vische, M. Cerebrospinal fluid (CSF) findings in amyotrophic lateral sclerosis. J. Neurol. 1984, 231, 75–78. [Google Scholar] [CrossRef]
  176. Meucci, G.; Rossi, G.; Bettini, R.; Montanaro, D.; Gironelli, L.; Voci, L.; Bianchi, F. Laser nephelometric evaluation of albumin, IgG and alpha 2-macroglobulin: Applications to the study of alterations of the blood-brain barrier. J. Neurol. Sci. 1993, 118, 73–78. [Google Scholar] [CrossRef]
  177. Pirttila, T.; Vanhatalo, S.; Turpeinen, U.; Riikonen, R. Cerebrospinal fluid insulin-like growth factor-1, insulin growth factor binding protein-2 or nitric oxide are not increased in MS or ALS. Acta Neurol. Scand. 2004, 109, 337–341. [Google Scholar] [CrossRef]
  178. Annunziata, P.; Volpi, N. High levels of C3c in the cerebrospinal fluid from amyotrophic lateral sclerosis patients. Acta Neurol. Scand. 1985, 72, 61–64. [Google Scholar] [CrossRef]
  179. Apostolski, S.; Nikolic, J.; Bugarski-Prokopljevic, C.; Miletic, V.; Pavlovic, S.; Filipovic, S. Serum and CSF immunological findings in ALS. Acta Neurol. Scand. 1991, 83, 96–98. [Google Scholar] [CrossRef]
  180. Brettschneider, J.; Petzold, A.; Sussmuth, S.D.; Ludolph, A.C.; Tumani, H. Axonal damage markers in cerebrospinal fluid are increased in ALS. Neurology 2006, 66, 852–856. [Google Scholar] [CrossRef]
  181. Chen, Y.; Liu, X.H.; Wu, J.J.; Ren, H.M.; Wang, J.; Ding, Z.T.; Jiang, Y.P. Proteomic analysis of cerebrospinal fluid in amyotrophic lateral sclerosis. Exp. Ther. Med. 2016, 11, 2095–2106. [Google Scholar] [CrossRef] [Green Version]
  182. Li, W.; Maeda, Y.; Yuan, R.R.; Elkabes, S.; Cook, S.; Dowling, P. Beneficial effect of erythropoietin on experimental allergic encephalomyelitis. Ann. Neurol. 2004, 56, 767–777. [Google Scholar] [CrossRef]
  183. Kim, Y.J.; Park, H.J.; Lee, G.; Bang, O.Y.; Ahn, Y.H.; Joe, E.; Kim, H.O.; Lee, P.H. Neuroprotective effects of human mesenchymal stem cells on dopaminergic neurons through anti-inflammatory action. Glia 2009, 57, 13–23. [Google Scholar] [CrossRef]
  184. Palmer, A.M. Multiple sclerosis and the blood-central nervous system barrier. Cardiovasc. Psychiatry Neurol. 2013, 2013, 530356. [Google Scholar] [CrossRef]
  185. Schellenberg, A.E.; Buist, R.; Yong, V.W.; Del Bigio, M.R.; Peeling, J. Magnetic resonance imaging of blood-spinal cord barrier disruption in mice with experimental autoimmune encephalomyelitis. Magn. Reson. Med. 2007, 58, 298–305. [Google Scholar] [CrossRef]
  186. Wu, F.; Cao, W.; Yang, Y.; Liu, A. Extensive infiltration of neutrophils in the acute phase of experimental autoimmune encephalomyelitis in C57BL/6 mice. Histochem. Cell Biol. 2010, 133, 313–322. [Google Scholar] [CrossRef]
  187. Liu, X.; Yao, D.L.; Webster, H. Insulin-like growth factor I treatment reduces clinical deficits and lesion severity in acute demyelinating experimental autoimmune encephalomyelitis. Mult. Scler. 1995, 1, 2–9. [Google Scholar] [CrossRef]
  188. Manouchehri, N.; Stuve, O. Choroid plexus volumetrics and brain inflammation in multiple sclerosis. Proc. Natl. Acad. Sci. USA 2021, 118. [Google Scholar] [CrossRef]
  189. Canavero, S.; Pagni, C.A.; Duca, S.; Bradac, G.B. Spinal intramedullary cavernous angiomas: A literature meta-analysis. Surg. Neurol. 1994, 41, 381–388. [Google Scholar] [CrossRef]
  190. El-Koussy, M.; Stepper, F.; Spreng, A.; Lukes, A.; Gralla, J.; Brekenfeld, C.; Sturzenegger, M.; Schroth, G. Incidence, clinical presentation and imaging findings of cavernous malformations of the CNS. A twenty-year experience. Swiss Med. Wkly. 2011, 141, w13172. [Google Scholar] [CrossRef]
  191. Gross, B.A.; Du, R.; Popp, A.J.; Day, A.L. Intramedullary spinal cord cavernous malformations. Neurosurg. Focus 2010, 29, E14. [Google Scholar] [CrossRef]
  192. McCormick, P.C.; Michelsen, W.J.; Post, K.D.; Carmel, P.W.; Stein, B.M. Cavernous malformations of the spinal cord. Neurosurgery 1988, 23, 459–463. [Google Scholar] [CrossRef]
  193. Mitha, A.P.; Turner, J.D.; Abla, A.A.; Vishteh, A.G.; Spetzler, R.F. Outcomes following resection of intramedullary spinal cord cavernous malformations: A 25-year experience. J. Neurosurg. Spine 2011, 14, 605–611. [Google Scholar] [CrossRef] [Green Version]
  194. Reitz, M.; Burkhardt, T.; Vettorazzi, E.; Raimund, F.; Fritzsche, E.; Schmidt, N.O.; Regelsberger, J.; Westphal, M.; Eicker, S.O. Intramedullary spinal cavernoma: Clinical presentation, microsurgical approach, and long-term outcome in a cohort of 48 patients. Neurosurg. Focus 2015, 39, E19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Anson, J.A.; Spetzler, R.F. Surgical resection of intramedullary spinal cord cavernous malformations. J. Neurosurg. 1993, 78, 446–451. [Google Scholar] [CrossRef] [PubMed]
  196. Khalatbari, M.R.; Hamidi, M.; Moharamzad, Y. Pediatric intramedullary cavernous malformation of the conus medullaris: Case report and review of the literature. Child’s Nerv Syst. 2011, 27, 507–511. [Google Scholar] [CrossRef] [PubMed]
  197. Wang, C.C.; Liu, A.; Zhang, J.T.; Sun, B.; Zhao, Y.L. Surgical management of brain-stem cavernous malformations: Report of 137 cases. Surg. Neurol. 2003, 59, 444–454, discussion 454. [Google Scholar] [CrossRef]
  198. Clatterbuck, R.E.; Eberhart, C.G.; Crain, B.J.; Rigamonti, D. Ultrastructural and immunocytochemical evidence that an incompetent blood-brain barrier is related to the pathophysiology of cavernous malformations. J. Neurol. Neurosurg. Psychiatry 2001, 71, 188–192. [Google Scholar] [CrossRef] [Green Version]
  199. Choi, J.P.; Wang, R.; Yang, X.; Wang, X.; Wang, L.; Ting, K.K.; Foley, M.; Cogger, V.; Yang, Z.; Liu, F.; et al. Ponatinib (AP24534) inhibits MEKK3-KLF signaling and prevents formation and progression of cerebral cavernous malformations. Sci. Adv. 2018, 4, eaau0731. [Google Scholar] [CrossRef] [Green Version]
  200. Davies, B.M.; Mowforth, O.D.; Smith, E.K.; Kotter, M.R. Degenerative cervical myelopathy. BMJ 2018, 360, k186. [Google Scholar] [CrossRef] [Green Version]
  201. Martin-Vaquero, P.; da Costa, R.C.; Allen, M.J.; Moore, S.A.; Keirsey, J.K.; Green, K.B. Proteomic analysis of cerebrospinal fluid in canine cervical spondylomyelopathy. Spine 2015, 40, 601–612. [Google Scholar] [CrossRef]
  202. Bartlett, R.D.; Eleftheriadou, D.; Evans, R.; Choi, D.; Phillips, J.B. Mechanical properties of the spinal cord and brain: Comparison with clinical-grade biomaterials for tissue engineering and regenerative medicine. Biomaterials 2020, 258, 120303. [Google Scholar] [CrossRef]
  203. Li, Y.Q.; Chen, P.; Haimovitz-Friedman, A.; Reilly, R.M.; Wong, C.S. Endothelial apoptosis initiates acute blood-brain barrier disruption after ionizing radiation. Cancer Res. 2003, 63, 5950–5956. [Google Scholar]
  204. Ljubimova, N.V.; Levitman, M.K.; Plotnikova, E.D.; Eidus, L. Endothelial cell population dynamics in rat brain after local irradiation. Br. J. Radiol. 1991, 64, 934–940. [Google Scholar] [CrossRef]
  205. Lossinsky, A.S.; Mossakowski, M.J.; Pluta, R.; Wisniewski, H.M. Intercellular adhesion molecule-1 (ICAM-1) upregulation in human brain tumors as an expression of increased blood-brain barrier permeability. Brain Pathol. 1995, 5, 339–344. [Google Scholar] [CrossRef]
  206. Carlos, T.M.; Clark, R.S.; Franicola-Higgins, D.; Schiding, J.K.; Kochanek, P.M. Expression of endothelial adhesion molecules and recruitment of neutrophils after traumatic brain injury in rats. J. Leukoc. Biol. 1997, 61, 279–285. [Google Scholar] [CrossRef]
  207. Clark, W.M.; Lauten, J.D.; Lessov, N.; Woodward, W.; Coull, B.M. Time course of ICAM-1 expression and leukocyte subset infiltration in rat forebrain ischemia. Mol. Chem. Neuropathol. 1995, 26, 213–230. [Google Scholar] [CrossRef]
  208. Nordal, R.A.; Wong, C.S. Intercellular adhesion molecule-1 and blood-spinal cord barrier disruption in central nervous system radiation injury. J. Neuropathol. Exp. Neurol. 2004, 63, 474–483. [Google Scholar] [CrossRef] [Green Version]
  209. Li, Y.Q.; Ballinger, J.R.; Nordal, R.A.; Su, Z.F.; Wong, C.S. Hypoxia in radiation-induced blood-spinal cord barrier breakdown. Cancer Res. 2001, 61, 3348–3354. [Google Scholar]
  210. Tsao, M.N.; Li, Y.Q.; Lu, G.; Xu, Y.; Wong, C.S. Upregulation of vascular endothelial growth factor is associated with radiation-induced blood-spinal cord barrier breakdown. J. Neuropathol. Exp. Neurol. 1999, 58, 1051–1060. [Google Scholar] [CrossRef] [Green Version]
  211. Piekarz, K.M.; Bhaskaran, S.; Sataranatarajan, K.; Street, K.; Premkumar, P.; Saunders, D.; Zalles, M.; Gulej, R.; Khademi, S.; Laurin, J.; et al. Molecular changes associated with spinal cord aging. Geroscience 2020, 42, 765–784. [Google Scholar] [CrossRef] [PubMed]
  212. Banks, W.A. The Blood-Brain Barrier Interface in Diabetes Mellitus: Dysfunctions, Mechanisms and Approaches to Treatment. Curr. Pharm. Des. 2020, 26, 1438–1447. [Google Scholar] [CrossRef]
  213. Cragg, J.J.; Noonan, V.K.; Dvorak, M.; Krassioukov, A.; Mancini, G.B.; Borisoff, J.F. Spinal cord injury and type 2 diabetes: Results from a population health survey. Neurology 2013, 81, 1864–1868. [Google Scholar] [CrossRef] [Green Version]
  214. Carrino, D.; Branca, J.J.V.; Becatti, M.; Paternostro, F.; Morucci, G.; Gulisano, M.; Di Cesare Mannelli, L.; Pacini, A. Alcohol-Induced Blood-Brain Barrier Impairment: An In Vitro Study. Int. J. Environ. Res. Public Health 2021, 18, 2683. [Google Scholar] [CrossRef]
  215. Wei, J.; Dai, Y.; Wen, W.; Li, J.; Ye, L.L.; Xu, S.; Duan, D.D. Blood-brain barrier integrity is the primary target of alcohol abuse. Chem. Biol. Interact. 2021, 337, 109400. [Google Scholar] [CrossRef]
  216. Valkov, T.; Hristova, J.; Tcherveniakova, T.; Svinarov, D. Blood-Brain Barrier and Intrathecal Immune Response in patients with neuroinfections. Infez. Med. 2017, 25, 320–325. [Google Scholar]
  217. Wang, Y.; Liu, X.; Liu, Q. NOD2 Expression in Streptococcus pneumoniae Meningitis and Its Influence on the Blood-Brain Barrier. Can. J. Infect. Dis. Med. Microbiol. 2018, 2018, 7292084. [Google Scholar] [CrossRef]
  218. Thompson, D.; Sorenson, J.; Greenmyer, J.; Brissette, C.A.; Watt, J.A. The Lyme disease bacterium, Borrelia burgdorferi, stimulates an inflammatory response in human choroid plexus epithelial cells. PLoS ONE 2020, 15, e0234993. [Google Scholar] [CrossRef]
  219. Carloni, S.; Bertocchi, A.; Mancinelli, S.; Bellini, M.; Erreni, M.; Borreca, A.; Braga, D.; Giugliano, S.; Mozzarelli, A.M.; Manganaro, D.; et al. Identification of a choroid plexus vascular barrier closing during intestinal inflammation. Science 2021, 374, 439–448. [Google Scholar] [CrossRef]
  220. Marques, F.; Sousa, J.C. The choroid plexus is modulated by various peripheral stimuli: Implications to diseases of the central nervous system. Front. Cell Neurosci. 2015, 9, 136. [Google Scholar] [CrossRef] [Green Version]
  221. Bertrand, L.; Cho, H.J.; Toborek, M. Blood-brain barrier pericytes as a target for HIV-1 infection. Brain 2019, 142, 502–511. [Google Scholar] [CrossRef] [PubMed]
  222. Rhea, E.M.; Logsdon, A.F.; Hansen, K.M.; Williams, L.M.; Reed, M.J.; Baumann, K.K.; Holden, S.J.; Raber, J.; Banks, W.A.; Erickson, M.A. The S1 protein of SARS-CoV-2 crosses the blood-brain barrier in mice. Nat. Neurosci. 2021, 24, 368–378. [Google Scholar] [CrossRef] [PubMed]
  223. Pellegrini, L.; Albecka, A.; Mallery, D.L.; Kellner, M.J.; Paul, D.; Carter, A.P.; James, L.C.; Lancaster, M.A. SARS-CoV-2 Infects the Brain Choroid Plexus and Disrupts the Blood-CSF Barrier in Human Brain Organoids. Cell Stem Cell 2020, 27, 951–961 e955. [Google Scholar] [CrossRef] [PubMed]
  224. Christie, M.J.; Irving, A.T.; Forster, S.C.; Marsland, B.J.; Hansbro, P.M.; Hertzog, P.J.; Nold-Petry, C.A.; Nold, M.F. Of bats and men: Immunomodulatory treatment options for COVID-19 guided by the immunopathology of SARS-CoV-2 infection. Sci. Immunol. 2021, 6, eabd0205. [Google Scholar] [CrossRef]
  225. Buzhdygan, T.P.; DeOre, B.J.; Baldwin-Leclair, A.; Bullock, T.A.; McGary, H.M.; Khan, J.A.; Razmpour, R.; Hale, J.F.; Galie, P.A.; Potula, R.; et al. The SARS-CoV-2 spike protein alters barrier function in 2D static and 3D microfluidic in-vitro models of the human blood-brain barrier. Neurobiol. Dis. 2020, 146, 105131. [Google Scholar] [CrossRef]
  226. Leda, A.R.; Bertrand, L.; Andras, I.E.; El-Hage, N.; Nair, M.; Toborek, M. Selective Disruption of the Blood-Brain Barrier by Zika Virus. Front. Microbiol. 2019, 10, 2158. [Google Scholar] [CrossRef]
  227. Shao, Q.; Herrlinger, S.; Yang, S.L.; Lai, F.; Moore, J.M.; Brindley, M.A.; Chen, J.F. Zika virus infection disrupts neurovascular development and results in postnatal microcephaly with brain damage. Development 2016, 143, 4127–4136. [Google Scholar] [CrossRef] [Green Version]
  228. Suwannasual, U.; Lucero, J.; Davis, G.; McDonald, J.D.; Lund, A.K. Mixed Vehicle Emissions Induces Angiotensin II and Cerebral Microvascular Angiotensin Receptor Expression in C57Bl/6 Mice and Promotes Alterations in Integrity in a Blood-Brain Barrier Coculture Model. Toxicol. Sci. 2019, 170, 525–535. [Google Scholar] [CrossRef]
  229. Yu, Y.; Wang, C.; Zhang, X.; Zhu, J.; Wang, L.; Ji, M.; Zhang, Z.; Ji, X.M.; Wang, S.L. Perfluorooctane sulfonate disrupts the blood brain barrier through the crosstalk between endothelial cells and astrocytes in mice. Environ. Pollut. 2020, 256, 113429. [Google Scholar] [CrossRef]
  230. Garate-Velez, L.; Escudero-Lourdes, C.; Salado-Leza, D.; Gonzalez-Sanchez, A.; Alvarado-Morales, I.; Bahena, D.; Labrada-Delgado, G.J.; Rodriguez-Lopez, J.L. Anthropogenic Iron Oxide Nanoparticles Induce Damage to Brain Microvascular Endothelial Cells Forming the Blood-Brain Barrier. J. Alzheimers Dis. 2020, 76, 1527–1539. [Google Scholar] [CrossRef]
  231. Miyazaki, W.; Fujiwara, Y.; Katoh, T. The effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on the development and function of the blood-brain barrier. Neurotoxicology 2016, 52, 64–71. [Google Scholar] [CrossRef]
  232. Calderon-Garciduenas, L.; Vojdani, A.; Blaurock-Busch, E.; Busch, Y.; Friedle, A.; Franco-Lira, M.; Sarathi-Mukherjee, P.; Martinez-Aguirre, X.; Park, S.B.; Torres-Jardon, R.; et al. Air pollution and children: Neural and tight junction antibodies and combustion metals, the role of barrier breakdown and brain immunity in neurodegeneration. J. Alzheimers Dis. 2015, 43, 1039–1058. [Google Scholar] [CrossRef]
  233. Rosell, A.; Cuadrado, E.; Ortega-Aznar, A.; Hernandez-Guillamon, M.; Lo, E.H.; Montaner, J. MMP-9-positive neutrophil infiltration is associated to blood-brain barrier breakdown and basal lamina type IV collagen degradation during hemorrhagic transformation after human ischemic stroke. Stroke 2008, 39, 1121–1126. [Google Scholar] [CrossRef] [Green Version]
  234. Ries, C. Cytokine functions of TIMP-1. Cell. Mol. Life Sci. 2014, 71, 659–672. [Google Scholar] [CrossRef]
  235. Li, D.D.; Song, J.N.; Huang, H.; Guo, X.Y.; An, J.Y.; Zhang, M.; Li, Y.; Sun, P.; Pang, H.G.; Zhao, Y.L.; et al. The roles of MMP-9/TIMP-1 in cerebral edema following experimental acute cerebral infarction in rats. Neurosci. Lett. 2013, 550, 168–172. [Google Scholar] [CrossRef]
  236. Tang, J.; Kang, Y.; Huang, L.; Wu, L.; Peng, Y. TIMP1 preserves the blood-brain barrier through interacting with CD63/integrin beta 1 complex and regulating downstream FAK/RhoA signaling. Acta Pharm. Sin. B. 2020, 10, 987–1003. [Google Scholar] [CrossRef]
  237. Souza, P.S.; Goncalves, E.D.; Pedroso, G.S.; Farias, H.R.; Junqueira, S.C.; Marcon, R.; Tuon, T.; Cola, M.; Silveira, P.C.L.; Santos, A.R.; et al. Physical Exercise Attenuates Experimental Autoimmune Encephalomyelitis by Inhibiting Peripheral Immune Response and Blood-Brain Barrier Disruption. Mol. Neurobiol. 2017, 54, 4723–4737. [Google Scholar] [CrossRef]
  238. Mokhtarzade, M.; Motl, R.; Negaresh, R.; Zimmer, P.; Khodadoost, M.; Baker, J.S.; Patel, D.; Majdinasab, N.; Ranjbar, R. Exercise-induced changes in neurotrophic factors and markers of blood-brain barrier permeability are moderated by weight status in multiple sclerosis. Neuropeptides 2018, 70, 93–100. [Google Scholar] [CrossRef] [Green Version]
  239. Kumarasamy, M.; Sosnik, A. Heterocellular spheroids of the neurovascular blood-brain barrier as a platform for personalized nanoneuromedicine. iScience 2021, 24, 102183. [Google Scholar] [CrossRef]
  240. Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L.; et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol. Ther. 2011, 19, 1769–1779. [Google Scholar] [CrossRef]
Figure 1. Blood-spinal cord barrier (BSCB) and its cellular components. (A) Cross section of spinal cord and its blood supply (B) Schematics of cellular components of BSCB; endothelial cells (ECs) are separated from pericytes by basement membrane and held together by tight junction proteins (TJs); astrocytes connect with both pericytes and ECs (C) Inset shows interaction between the astrocytic end foot enriched with aquaporin 4 (AQP4) and ECs in presence of immune cells (microglia) (D) Inset shows TJs like endothelial cell-selective adhesion molecule (ESAM), claudins, occludins and Zonula occludens (ZO-1) holding the ECs together (E) Inset represents interaction between ECs and pericytes (expressing PDGFR and CD13) that plays an important role in maintaining the integrity of BSCB by regulating the uptake of circulating macromolecules (presented as yellow sign).
Figure 1. Blood-spinal cord barrier (BSCB) and its cellular components. (A) Cross section of spinal cord and its blood supply (B) Schematics of cellular components of BSCB; endothelial cells (ECs) are separated from pericytes by basement membrane and held together by tight junction proteins (TJs); astrocytes connect with both pericytes and ECs (C) Inset shows interaction between the astrocytic end foot enriched with aquaporin 4 (AQP4) and ECs in presence of immune cells (microglia) (D) Inset shows TJs like endothelial cell-selective adhesion molecule (ESAM), claudins, occludins and Zonula occludens (ZO-1) holding the ECs together (E) Inset represents interaction between ECs and pericytes (expressing PDGFR and CD13) that plays an important role in maintaining the integrity of BSCB by regulating the uptake of circulating macromolecules (presented as yellow sign).
Neurosci 03 00001 g001
Table 1. Key differences between BBB and BSCB.
Table 1. Key differences between BBB and BSCB.
FeatureBlood-Brain Barrier (BBB)Blood-Spinal Cord Barrier (BSCB)References
PermeabilityLowHigh: 3H-mannitol and 14C-inulin[19]
Tight Junction proteinsHighLow: ZO-1, occludin, β-catenin, VE-cadherin[20]
Number of pericytesHighLow[21]
Glycogen DepositsLowHigh[7]
Table 2. Implications of blood-spinal cord barrier dysfunction in different spinal cord disorders.
Table 2. Implications of blood-spinal cord barrier dysfunction in different spinal cord disorders.
CellsCause/Effects of BSCB ImpairmentRefs.
Spinal Cord Injury (SCI)
MicrogliaJmjd3 ↑ → NF-κB → MMP3 ↑ and MMP9 ↑[44]
TRPV4 ↑ → spinal cord scarring, endothelial damage, BSCB damage[47]
MMP-8 ↑ → occludin ↓ and ZO-1 ↓[45]
MMP-12 → functional recovery↓, BSCB permeability ↓[46]
USP4 ↓; NF-κB → TRFAF6 → Neuronal inflammation[48]
AR deficiency → M2 response → locomotion recovery
AR inhibition → HNE accumulation → phosphorylation of CREB → Arg1‪ ↑
[49]
AQP4 ↑ → BSCB permeability ↑[50]
Reactive astrocytes (RAs)Shh/Gli ↑ → BSCB permeability ↑, locomotor recovery ↓[25]
Ras → fibronectin/β1 integrin pathway → microglial inflammation[51]
Calmodulin → AQP4 ↑[52]
MacrophagesPerforin ↑ → BSCB permeability ↑ → cytokine and inflammatory cell infiltration[49,51,53]
NeutrophilsMMP-3 ↑ → NF-κB → occludin ↓ and ZO-1 ↓[43]
Amyotrophic Lateral Sclerosis (ALS)
AstrocytesWnt7a ↓, Wnt5a ↓
Gi signalling in astrocytes restores BSCB integrity
[54]
Swollen astrocyte foot processes
Degenerating astrocytes
[55]
Glutamate ↑ → EC P-gp ↑, NMDA ↑[56]
NeuronsMotor neuron loss[9,30,35,54,55,56,57,58,59,60,61,62,63]
PDGFC ↑ → BSCB dysfunction [57]
Immune cellsErythrocyte extravasation[30]
PericyteReduction in pericytes[30,35]
Endothelial cellsGlut-1 ↓, CD146 ↓[9]
Claudin 5 ↓, occludin ↓, ZO-1↓[60,62,63]
Cytoplasmic vacuoles[61]
Mitochondrial degeneration[55]
P-gp ↑, BCRP ↑, MRP2 ↑[56,57,59]
ROS ↑[62]
Circulating ECs ↓[58]
ECMAgrin ↓[60]
Peripheral Nerve Injury (PNI)
MicrogliaMCP-1 ↑; EB extravasation ↑; IL-1β ↑; TGF-β1 ↑; ZO-1, occludin ↓[64]
MCP-1 → microglial activation → neuropathic pain → delayed astrocyte activation[65]
AstrocytesAQP4 ↑ → length and volume of astrocytic processes ↑[66]
SUR1-TRPM4 ↑ → dorsal horn astrocytes[67]
Degenerative Cervical Myelopathy (DCM)
Immune cellsAngiopoietin 2 ↓, VEGF C ↓[68]
Peripheral monocytes ↑[69]
IgGA ↑, IgGQ ↑, BSCB permeability ↑[29]
Multiple Sclerosis
Endothelial cellsClaudin-11 ↓; BSCB permeability ↑[70]
MicrogliaTMP → STAT3/SOC3 → NF-κB → M1 to M2 polarization
TNF-α ↑, IL-1β ↑, IL-4 ↓, IL-10 ↓
[26]
NeutrophilsIL-R1 → adhesion of neutrophils to inflamed SC[71]
Spinal Cord Ischemia
MicrogliaTLR4/MyD88/TRIF ↓
Inflammation ↓
[72]
HMGB1 ↑[73]
CXCL13/CXCR5 ↑ → ERK[74]
TUG ↓ → TRIL ↓ → NF-κB/IL-1β ↓[75]
Nrf2 ↑ → p53/p38/MAPK/NF-κB → ABC transporters[76]
Cancer
MicrogliaZO-1 ↓, claudin-5 ↓[77]
PDGF = Platelet-derived growth factor; MRP2 = Multidrug resistance protein; EC = Endothelial cell; NMDA = N-methyl-D-aspartic acid; BCRP = Breast cancer resistance protein; ZO = Zona occludens; NF-κB = Nuclear factor kappa B; AR = Aldose reductase; HNE = 4-hydroxynonenal; RAs = Reactive astrocytes; TMP = Tetramethylpyrazine; CREB =cAMP response element-binding protein; TUG = Taurine-upregulated gene 1; HMGB1 = high-mobility group box-1; TLR4 = Toll-like receptor 4; TRIL = TLR4 interactor with leucine-rich repeats; MMP = Matrix metalloproteinases; BSCB = Blood-spinal cord barrier; MCP-1 = Monocyte Chemoattractant Protein-1; SC = Spinal cord; TLR4 = Toll-like receptors 4; MyD88 = Myeloid differentiation factor 88; TRIF = TIR domain-containing adaptor inducing IFN-β.
Table 3. Therapeutic options for blood-spinal cord barrier in different spinal cord disorders.
Table 3. Therapeutic options for blood-spinal cord barrier in different spinal cord disorders.
InterventionMechanismsRefs.
DRUGSSpinal Cord Injury (SCI)
Valproic acidM2 polarization
HDAC3, STAT1 ↓
TNF-α, IL-1β, IL-6, IFN-γ ↓
[87]
DL-3-n-butylphthalide (DL-NBP)Motor function ↑, oedema ↓[90]
Bradykinin B2 receptor antagonist- HOE-140SC blood flow ↑; nNOS ↑
BSCB disruption ↓, oedema formation ↓
[91]
Protocatechuic acid (PCA)Apoptotic cell death of neurons and oligodendrocytes ↓
Infiltration of neutrophils and macrophages ↓
MMP-9 ↓
TNFα, IL-1β, cyclooxygenase-2, inducible nitric oxide synthase and Chemokines ↓
[83]
Brilliant blue G (BBG)P2X7, NLRP3, ASC, cleaved XIAP, caspase-1, caspase-11, IL-1ß, IL-18 ↑[92]
Human immunoglobulin G (hIgG)Antagonize neutrophil infiltration
Neutrophil chemo-attractants ↑
[93,94]
Haem oxygenase (HO)-14-Hydroxynonenal (4-HNE), malondialdehyde (MDA) ↑[95]
NaHS
(H2S donor)
TJ proteins ↑
BSCB permeability ↓
[96]
Gallic acid (GA)Jmjd3 ↓, MMP9 ↓
Neutrophil and macrophage infiltration ↓
[97]
Dl-3-n-butylphthalide (NBP)ER stress ↓
Occludin, p120-Catenin, β-Catenin, claudin-5 ↑
[98]
Lithium chloride (LiCl)Occludin, claudin-5 ↑
ER stress ↓
LC3-II, ATG-5 ↑ p62 ↓
[99,100]
LycopeneWater content ↓
TNF-α and NF-kB ↓
ZO-1, claudin-5 ↑
[101]
Phenylbutyrate (PBA)p120, β-catenin, occludin, claudin5 ↑
ER stress ↓
BSCB permeability ↓
[102,103]
Bromodomain and extra-terminal domain (BET) proteinsPro-inflammatory mediators ↓
Anti-inflammatory cytokines ↑
Reactivity of microglia/macrophages ↓
Neuroprotection and functional recovery ↑
[104]
Folic acid (FA)MMP2 ↓[105]
Flufenamic acidTrpM4 ↓
MMP2 ↓, MMP9 ↓
[84]
Valproic acidMicroglia polarization; ↓TNF-α, IL-1β, IL-6, INF-γ[87]
Oleanolic acid (OA)Caspase-3 ↓
Pro-inflammatory response ↓
MAPKs, NF-κB ↓
[88]
Salvianolic acid (A and B)ZO-1, occludin ↑
TNF-α and NF-κB ↓
miR-101/Cul3/Nrf2/HO-1 ↑
[89,106]
Fluoxetine MMP2 ↓, MMP9 ↓
ZO-1, occludin ↑
Groα, MIP1α and 1β ↓
Infiltration of neutrophils and macrophage ↓
[107,108]
Simvastatin-ezetimibeICAM-1 ↓
Endothelial inflammatory response ↓
Wnt/β-catenin ↑
[109,110]
Retinoic acid (RA)P120, β-catenin, occludin and claudin5 ↑
CHOP ↓, caspase12 ↓
[111]
Epidermal growth factor (EGF)Bax ↓, Bcl-2 ↑
Superoxide dismutase (SOD) ↑ glutathione peroxidase (GPx) ↑
[112]
Basic fibroblast growth factor (bFGF)MMP9 ↓
Caveolin-1, TJs, including occludin, claudin-5, p120-catenin and β-catenin ↑
PRDX1 ↑→ autophagy
Neuroprotection ↑, axonal regeneration
[113,114]
Methylprednisolone (MP) and aminoguanidine (AG)AQP4 ↓
iNOS ↓
[115,116]
Tetramethylpyrazine (TMP)BSCB permeability ↓
IL-1β, TNFα, IL-18, TUNEL-positive cells, caspase 3/9 ↓
[117]
CurcuminTNF-α and NF-κB ↓
ZO-1, occludin ↑
[118]
EugenolNF-κB, p38 MAPK ↓
Inflammation, oxidative stress ↓
[119]
Mithramycin A (MA) Ghrelin
17β-estradiol (E2)
MMP9 ↓ SUR1/TRPM4 ↓
TJs ↑
[81,120]
NANOPARTICLESCarbon monoxide-releasing molecule-2 (CORM-2)ZO-1, ZO-2, occludin and claudin-1 ↑ BSCB permeability ↓[121]
CORM-3TJs ↑, MMP9 ↓[122]
Bone mesenchymal stem cell-derived extracellular vesicles (BMSC-EV)Brain cell death ↓
Neuronal survival ↑, motor function ↑
Pericyte migration ↓
[123]
Poly (d,l-lactide co-glycolide, PLGA)-based NPsLocalization at lesion site[124,125,126]
BIOMATERIALSAstragoloside IV Loaded Polycaprolactone MembraneCaspase3 ↓, Bax/Bcl-2 ↓
Occludin, claudin5, ZO-1 ↑
MMP9 ↓, neutrophil infiltration ↓
BSCB permeability ↓
[127]
MSCsBSCB leakage ↓
von Willebrand factor (vWF) ↑
Locomotor function ↑
[128]
HAMC/PLGA/FGF2FGF2 ↑[129]
miRNAsmiRNA-125a-5pZO-1, occludin, VE-cadherin ↑
BSCB permeability ↓
[130]
miR-429ZO-1, occludin and claudin-5 ↑
Krüppel-like factor 6 (KLF6) ↓
[131,132]
Ad-GFP-HO-1C[INCREMENT]23Hindlimb function ↑
TJs ↑
[133]
Amyotrophic Lateral Sclerosis (ALS)
DRUGSAPC (Activated protein C)IgG and iron deposition ↓
ZO-1, occludin ↑
[134,135]
AMD3100 CXCR4/CXCL12 ↓
Microglial pathology ↑
Proinflammatory cytokines ↓
[136]
BIOMATERIALSUnmodified human bone marrow CD34+ (hBM34+) stem cellsEB extravasation ↓
Restored capillary ultrastructure
Engrafted widely into capillaries of the gray/white matter SC and brain Motor cortex/brainstem
structural and functional repair of BSCB impairment
[137,138,139]
Human bone marrow-derived endothelial progenitor cells (hBMEPCs)VEGF-A and angiogenin-1 ↑
EC phenotype ↑
ZO-1, occludin ↑
[140]
Mesenchymal stem cells (MSCs)Motor neuron loss ↓
Locomotor activity ↑
Neurturin ↑
[141]
Spinal Cord Ischemia
DRUGSPropofolBSCB permeability ↓, MMP9 ↓, NF-kB ↓
Occludin ↑, claudin-5 ↑
[142]
Dexmedetomidine (Dex)HMGB1-TLR4-NF-κB signalling pathway ↓
MMP9 ↓, angiopoietin-1 (Ang1) and Tie2 ↑
[143,144]
Remote ischemic preconditioning (RIPC)Cannabinoid-1,2 receptors ↑
BSCB integrity ↑
ZO-1 ↑
MMP9 ↓, TNF-α ↓
[145,146]
SevofluraneMMP9 ↓
CXCL10, CCL2 ↓
IL-1β ↓
[147]
BIOMATERIALSBM-MSCsEB extravasation ↓
MMP9 ↓, TNF-α ↓
[148,149]
miRNAmiR-128-3pSpecificity protein 1 (SP1) ↓
Neuroinflammation ↓
[150]
miR-320aAQP1 ↓[151]
miR-27aTICAM-2 ↓→NF-κB ↓[152]
Multiple Sclerosis
DRUGSTetramethylpyrazine (TMP)TNF-α, IL-1β ↓
IL-4, IL-10 ↑; TJs ↑
STAT3/SOCS3 ↑→NF-кB ↓→M2 polarization
[117]
Calcitriol (vitamin D analog)NLRP3, caspase-1, (IL)-1β, CX3CR1, CCL17, RORc, Tbx21 ↓
MHCII ↓
ZO-1 ↑
[153]
ADAMTS13VWF ↓
Demyelination ↓
T lymphocyte, neutrophil and monocyte infiltration ↓
[154]
Glyceryl tribenzoate (GTB) and
Cinnamon
Perivascular cuffing ↓
Inflammation ↓
TGF-β, regulatory T cells (Tregs) in splenocytes ↑
[155,156]
BIOMATERIALSMSCs-IFN-β+minocyclineIFN-γ, TNF-α ↓
IL-4, IL-10 ↑
MMP9 ↓
[157]
Peripheral Nerve Injury (PNI)
Salmon thrombinTNF-α-induced endothelial permeability ↓
BSCB breakdown ↓
[158]
HDACs: Histone deacetylases; STATs: Signal transducers and activators of transcriptions; TNF: Tumour necrosis factor; IL: Interleukin; IFN: Interferon; nNOS: nitric oxide synthase; MMPs: Matrix metalloproteinases; NLR: NOD-like receptor; ASC: apoptosis-associated speck-like protein containing CARD; JMJD3:Jumonji domain-containing protein D3; ER: Endoplasmic reticulum; LC3: Microtubule-associated protein light chain 3; ATG5: Autophagy related 5; ZO-1: Zonula occludens-1; TrpM4: transient receptor potential cation channel subfamily M member 4; NF-κB: Nuclear factor-κB; MAPK4: mitogen-activated protein kinase; HO-1: heme oxygenase-1; Cul3:Cullin 3; Nrf2: Nuclear factor-erythroid factor 2-related factor 2; GROα:Growth-regulated oncogene α; MIP1α:macrophage inflammatory proteins; ICAM-1: Intercellular Adhesion Molecule 1; CHOP: CCAAT-enhancer-binding protein homologous protein; PRDX1:Peroxiredoxin 1; AQP4: Aquaporin 4; IgG: Immunoglobulin; CXCR: C-X-C chemokine receptor type; VEGF: Vascular endothelial growth factor; SOCS3:Suppressor Of Cytokine Signalling 3; RORC:RAR Related Orphan Receptor C; MHC: major histocompatibility complex.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chopra, N.; Menounos, S.; Choi, J.P.; Hansbro, P.M.; Diwan, A.D.; Das, A. Blood-Spinal Cord Barrier: Its Role in Spinal Disorders and Emerging Therapeutic Strategies. NeuroSci 2022, 3, 1-27. https://doi.org/10.3390/neurosci3010001

AMA Style

Chopra N, Menounos S, Choi JP, Hansbro PM, Diwan AD, Das A. Blood-Spinal Cord Barrier: Its Role in Spinal Disorders and Emerging Therapeutic Strategies. NeuroSci. 2022; 3(1):1-27. https://doi.org/10.3390/neurosci3010001

Chicago/Turabian Style

Chopra, Neha, Spiro Menounos, Jaesung P. Choi, Philip M. Hansbro, Ashish D. Diwan, and Abhirup Das. 2022. "Blood-Spinal Cord Barrier: Its Role in Spinal Disorders and Emerging Therapeutic Strategies" NeuroSci 3, no. 1: 1-27. https://doi.org/10.3390/neurosci3010001

Article Metrics

Back to TopTop