Next Article in Journal
E7050 Suppresses the Growth of Multidrug-Resistant Human Uterine Sarcoma by Inhibiting Angiogenesis via Targeting of VEGFR2-Mediated Signaling Pathways
Next Article in Special Issue
Pharmacokinetics and Tissue Distribution of Bee Venom-Derived Phospholipase A2 Using a Sandwich ELISA after Subcutaneous Injection of New Composition Bee Venom in Rats
Previous Article in Journal
Sleep Deprivation and Gut Microbiota Dysbiosis: Current Understandings and Implications
Previous Article in Special Issue
Depletion of Arg1-Positive Microglia/Macrophages Exacerbates Cerebral Ischemic Damage by Facilitating the Inflammatory Response
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The S100B Protein: A Multifaceted Pathogenic Factor More Than a Biomarker

by
Fabrizio Michetti
1,2,3,4,*,
Maria Elisabetta Clementi
5,
Rosa Di Liddo
6,
Federica Valeriani
7,
Francesco Ria
8,
Mario Rende
9,
Gabriele Di Sante
9 and
Vincenzo Romano Spica
7
1
Department of Neuroscience, Catholic University of the Sacred Heart, 00168 Rome, Italy
2
IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, 20132 Milan, Italy
3
Department of Medicine, LUM University, 70010 Casamassima, Italy
4
Genes, Via Venti Settembre 118, 00187 Roma, Italy
5
Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” SCITEC-CNR, 00168 Rome, Italy
6
Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
7
Laboratory of Epidemiology and Biotechnologies, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy
8
Department of Translational Medicine and Surgery, Section of General Pathology, Catholic University of the Sacred Heart, 00168 Rome, Italy
9
Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 06132 Perugia, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9605; https://doi.org/10.3390/ijms24119605
Submission received: 26 April 2023 / Revised: 29 May 2023 / Accepted: 30 May 2023 / Published: 31 May 2023
(This article belongs to the Special Issue Neuroinflammatory Processes in Neurodegenerative Diseases 2.0)

Abstract

:
S100B is a calcium-binding protein mainly concentrated in astrocytes in the nervous system. Its levels in biological fluids are recognized as a reliable biomarker of active neural distress, and more recently, mounting evidence points to S100B as a Damage-Associated Molecular Pattern molecule, which, at high concentration, triggers tissue reactions to damage. S100B levels and/or distribution in the nervous tissue of patients and/or experimental models of different neural disorders, for which the protein is used as a biomarker, are directly related to the progress of the disease. In addition, in experimental models of diseases such as Alzheimer’s and Parkinson’s diseases, amyotrophic lateral sclerosis, multiple sclerosis, traumatic and vascular acute neural injury, epilepsy, and inflammatory bowel disease, alteration of S100B levels correlates with the occurrence of clinical and/or toxic parameters. In general, overexpression/administration of S100B worsens the clinical presentation, whereas deletion/inactivation of the protein contributes to the amelioration of the symptoms. Thus, the S100B protein may be proposed as a common pathogenic factor in different disorders, sharing different symptoms and etiologies but appearing to share some common pathogenic processes reasonably attributable to neuroinflammation.

1. Introduction

The term S100B refers to a protein identified in the mid-1960s from brain extracts to compare proteins identified in the nervous tissue with those localized in other tissues. It is characterized by an intriguing, unusual solubility in a 100% saturated solution with ammonium sulfate [1], and this characteristic was the basis of its denomination, which was originally S100 protein. At present, the S100 protein family comprises more than 20 calcium-binding proteins, mostly formed by two identical peptides (homodimers), exhibiting structural similarities with different degrees of amino acid homology, located in different tissues where they modulate the activity of many targets [2]. S100B is an acidic homodimer of 9–14 kDa per peptide (monomer) and constitutes the bulk of the protein fraction that was originally isolated from brain extracts; thus, it has been regarded as being specific for this tissue for approximately two decades, but later was shown also to be present in definite extra-neural cell types, as indicated below. Interestingly, the amino acid composition and conformation of S100B, as for other proteins of the S100 family, are highly conserved in vertebrate species, suggesting that it may have a crucially conserved biological role(s), although they have not been identified. In the nervous system, although S100B is mainly concentrated in astrocytes, it is also expressed in other glial cell types, such as oligodendrocytes, Schwann cells, ependymal cells, retinal Müller cells, and enteric glial cells. The protein has even been reported to be contained in specific neuronal subpopulations in the brainstem and some ganglionic peripheral cells. In addition, as above indicated, S100B has also been detected in definite non-neural cell types, such as chondrocytes, melanocytes, Langerhans cells, dendritic cells of lymphoid organs, some lymphocyte cell types, adrenal medulla satellite cells, skeletal muscle satellite cells, tubular kidney cells, non-nervous structures of the eye, such as corneal endothelial cells and lens, iris, ciliary body epithelial cells, Leydig cells, and adipocytes, which constitute a site of concentration for the protein comparable to astrocytes [3]. While the functional characteristics and cell localization of neural S100B have been extensively studied, its properties in non-neural locations have received poor attention, although they would reasonably deserve analogous consideration. These risks becoming a gap in studies concerning the role of S100B in physiological and/or pathological conditions. In any case, with this potential problem in mind, this review will focus on the involvement of S100B in diseases of the nervous system, leaving out extra neural disorders such as obesity, diabetes, and melanoma. Indeed, the cell distribution of this protein does not offer conclusive clues to its functional role(s). In general terms, S100B, such as other calcium-binding proteins belonging to the S100 family, appears to regulate a variety of intracellular activities, interacting with different molecules located in different cell types [4]. The different functions attributed to the protein, which include the regulation of cellular calcium homeostasis and enzyme activities, interaction with the cytoskeleton, cell survival, cell differentiation, and cell proliferation, do not appear to delineate a clear, univocal intracellular role for S100B.
In contrast, growing evidence indicates an increasingly clearer role for S100B when it is secreted—reasonably mostly by astrocytes in the nervous system—in the extracellular compartment. Extracellular S100B is regarded as interacting with target cells mainly, but not exclusively, through the muti-ligand transmembrane Receptor for Advanced Glycation End Products (RAGE), initiating intracellular signaling cascades that may result in physiological regulation at low nanomolar concentrations (“Jekyll side”) or various pathological conditions, acting as a Danger/Damage Associated Molecular Pattern (DAMP) protein at higher micromolar concentrations (“Hyde side”) [5]. RAGE is regarded as a pattern recognition receptor capable of recognizing molecules detectable in pathological conditions, such as DAMPs. The activation of the extracellular RAGE domain, in particular the V domain, which is located at the most lateral position from the plasma membrane, activates transcription factors such as NF-κB, leading to increased expression of proinflammatory cytokines [6]. Following the synthesis of the molecule, post-translational modifications induced by the interaction of extracellular S100B with its receptor are currently under active investigation to tailor an adequate modulation of these phenomena [7]. Thus, increased levels of S100B have also been shown in different biological fluids (cerebrospinal fluid, peripheral and cord blood, amniotic fluid, saliva, urine, and feces) during various pathological conditions involving the nervous system (neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), traumatic and vascular acute brain injury, epilepsy, and also inflammatory bowel disease, perinatal neural disorders, glioma, and psychiatric disorders) [8,9], but also extra-neural districts (obesity and diabetes, melanoma) [3]. Although the wide spectrum of diseases in which the protein is involved reduces its specificity, levels of S100B protein in biological fluids are recognized as an important aid in monitoring the trend of the disorder, also in consequence of therapeutic approaches, and constitute a reliable, even predictive, biomarker of active distress [5]. Interestingly, in this respect, serum S100B levels have also been shown to constitute a marker of severity in COVID-19 patients [10].
In this review, while the role of S100B as a reliable biomarker is essentially generally accepted, we focus on the more recently emerging evidence individuating the S100B protein as a pathogenic factor potentially involved in processes caused by different etiologic factors and displaying different symptoms, possibly sharing aspects attributable to inflammation. This is essentially based on results obtained in experimental models of different diseases and on the significant effects obtained by modulating this protein (Figure 1). Of course, data from humans will be needed to validate S100B for clinical use. The following sections of this review will show alterations of S100B in different disorders (AD, PD, ALS, MS, traumatic and vascular acute brain injury, epilepsy, and inflammatory bowel disease), as well as indicate how alterations of the protein in experimental models of disease correlate with clinical symptoms and/or pathological parameters.

1.1. S100B in Alzheimer’s Disease

Astrocytes, which are known to be the main site of concentration for S100B in the central nervous system, are also known to be its main homeostatic regulator; in AD, they exert both neuroprotective and neurotoxic effects depending on the disease stage and microenvironmental factors. Essentially, based on their involvement in neuroinflammatory processes through the activation of intracellular pathways and the release of proinflammatory cytokines, they are believed to actively participate in AD pathogenic processes [11]. Astrocytic S100B has been shown to be upregulated in tissues of AD patients, and its abnormal levels, as a neurotrophic factor, have also been regarded as one possible explanation for the increased concentration of aggregates of overgrown neurites in the neuritic plaques [12,13,14,15,16]. S100B in biological fluids has been regarded as a reliable biomarker for the disease [17]. In particular, S100B levels in cerebrospinal fluid, together with other AD biomarkers such as amyloid β and phosphorylated τ, have recently been shown to have distinctive associations with higher gray matter volumes and increased glucose metabolism in key Alzheimer-related regions [18]. In fact, experiments using AD animal models indicated a significant role for the protein in AD pathogenic processes. When transgenic mice overexpressing S100B (TghuS100B mice) were crossed with the Tg2576 mouse model of AD, brain parenchymal and cerebral vascular β-amyloid (Aβ) deposits and Aβ levels were increased, accompanied by reactive astrocytosis and microgliosis and increased production of inflammatory cytokines [19]. Likewise, Aβ42 levels were significantly increased in the hippocampus and frontal cortex of transgenic mice overexpressing S100B, also exhibiting, interestingly, a sex-dependent manner [20]. On the contrary, inhibition of the protein ameliorated clinical conditions and pathological parameters in animal experimental models of the diseases. In particular, when pentamidine (PTM), an antiprotozoal drug that blocks S100B action, was administered to the mouse model of AD obtained using Aβ 1–42, reduced neuronal loss and gliosis were observed [21]. Likewise, the inhibition of astrocytic S100B synthesis by administering arundic acid (AA) reduced Aβ and amyloid plaque-associated gliosis in transgenic mice overproducing mutant amyloid precursor protein [22]. Additionally, genetic ablation of S100B (S100B knockout mice) resulted in reduced astrocytosis, microglia, dystrophic neurons, and plaques in animals generated by crossing transgenic AD model males with S100B knockout females [23]. In addition, gene polymorphisms upregulating S100B expression were interestingly shown to be associated with an increase in AD risk [24]. However, in vitro data indicating that S100B multimers act as complementary suppressors of Aβ42 oligomerization and aggregation, underpinning their potential neuroprotective role in AD, have also been reported, with the limitations of the mere in vitro approach [25].

1.2. S100B in Parkinson’s Disease

S100B has been reported to be overexpressed in the brain tissue of PD patients [26], as well as in animal PD models obtained using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) [27,28]. In addition, it has been demonstrated that mice overexpressing S100B are prone to developing Parkinsonian features [29] and that the exposure of midbrain cultures to S100B has been reported to specifically alter the activity of tyrosine hydroxylase-expressing (TH+) dopaminergic neurons [30]. Interestingly, overnight S100B elevation has been shown to correlate with increased PD severity and sleep disruption [31], while S100B polymorphisms appear to be associated with the age of onset of PD [32]. The use of S100B as a biomarker in biological fluid for monitoring the disease has also been considered based on results obtained in PD patients [33].
Additionally, in the PD experimental animal model, the administration of the inhibitor of S100B activity, PTM, resulted in a significant amelioration of motor performance [34]. Likewise, the inhibition of astrocytic S100B synthesis resulting from AA administration induced the protection of dopaminergic neurons in MPTP-treated mice as an experimental PD animal model [35]. A crucial role of S100B pathogenic processes was confirmed in S100B knockout mice treated with MPTP to induce PD, where the lack of S100B expression was accompanied by amelioration of pathological parameters such as reduced loss of dopaminergic neurons, reduced microgliosis, and reduced expression of tumor necrosis factor (TNF) alpha [27]. The role of S100B in PD pathogenesis has also been recently delineated in a comprehensive review [33].

1.3. S100B in Amyotrophic Lateral Sclerosis

High levels of S100B protein have been described in neural tissues, especially in astrocytes, from ALS patients [36,37], although conflicting data have been reported about the reliability of S100B as a biomarker in biological fluids for this disease [38,39,40]. S100B has also been reported to be overexpressed, together with its receptor RAGE and High Mobility Group Box 1 (HMGB1) protein, another DAMP molecule binding RAGE, in the lumbar spinal cord in the SOD-G93A rodent model for ALS. Thus, a potential role for these molecules in the progression of ALS has been further proposed [41]. Interestingly, a subpopulation of “aberrant” astrocytes overexpressing S100B and its receptor RAGE has been delineated as potentially involved in neuron toxicity, at least in the SOD-G93A animal experimental model of ALS [42,43]. However, correlations between pathological parameters of ALS and S100B levels at present are not available in vivo but only in cultured astrocytic cells. The silencing of S100B in astrocytes derived from the SOD1-G93A mouse model inhibited several genes commonly overexpressed in ALS astrocytes (TNF-α, C-X-C motif chemokine, chemokine (C-C motif) ligand 6, Glial Fibrillary Acidic Protein). Consistently, in the C6 rat astrocytoma cell line, S100B was overexpressed and extracellularly released at high levels when the SOD1-G93A mutated gene, responsible for experimental ALS induction, was transfected and transiently overexpressed [43]. Taken together, these results propose a role for S100B in ALS pathogenic processes. However, this putative pathogenic role of S100B in ALS needs to be confirmed by in vivo experiments.

1.4. S100B in Multiple Sclerosis

Earlier data correlating S100B to MS were obtained in the late 1970s, when high levels of the protein were detected in the cerebrospinal fluid (CSF) of MS patients during the acute phase, while levels were normal in the remission phase [44]. These results were confirmed after many years, and, in addition, S100B protein was also shown to appear at high levels in the serum of MS patients at onset [45]. The occurrence of S100B protein in the biological fluids of MS patients was found to correspond to features of the nervous tissue of MS patients. In post mortem tissues from these patients, an increased expression of S100B was detected both in active demyelinating and in chronic active MS plaques [46], and, in addition, RAGE has been shown to be overexpressed in active demyelinating lesions [45]. These data proposing the involvement of the S100B protein in MS were confirmed in experimental models of the disease. Interestingly, in a rodent demyelinating model of MS (experimental autoimmune encephalomyelitis—EAE), blockade of the S100B receptor RAGE was shown to suppress demyelination [47]. Inherently, the S100B/RAGE axis has later been shown to play a crucial role in myelination processes in oligodendrocyte cultures [48]. In EAE mice, inhibition of S100B activity using PTM [49] induced amelioration of clinical scores coherently accompanied by amelioration of pathological/biomolecular parameters, thus offering in vivo evidence that S100B plays a crucial role in pathogenic processes of MS. Consistent results were obtained after inhibition of astrocytic S100B synthesis by administering AA [50]. Finally, additional results confirming that S100B inhibition in vivo, using PTM or genetic ablation, protects from EAE were later obtained [51].

1.5. S100B in Traumatic and Vascular Acute Neural Injury

S100B levels in biological fluids constitute a recognized clinical parameter to evaluate patients with acute brain injury [52,53,54,55,56,57,58,59,60], and a special emphasis has been recently placed on mild traumatic brain injury [61,62,63,64,65,66,67,68,69,70,71] since S100B levels have been proposed as a reliable screening tool in this pathological condition. S100B levels in human brain tissue and their direct correlation with S100B in biological fluids are difficult to evaluate. This difficulty is mainly related to the permeability of the blood-brain barrier and the presence of S100B in extra-neural districts, this latter aspect being especially relevant in multiple trauma patients [9,72,73,74]. However, reasonably high S100B levels in biological fluids are, at least in large part, due to S100B levels and its release in brain tissue. S100B serum levels have also been proposed as a biomarker to distinguish patients with primary headaches from patients with secondary headaches, depending on other brain pathological conditions [75]. It should be noted that the use of S100B levels in biological fluids in order to monitor the effects of acute brain injury, also in light of official guidelines for this purpose, constitutes a topic that has been especially addressed in recent years. In this respect, the use of S100B as a valuable biomarker has also been compared with the other frequently used astrocytic biomarker, Glial Fibrillary Acidic Protein (GFAP) [9,63,68,69]. In fact, the use of S100B as a screening tool, especially in mild traumatic brain injury, offered potential advantages over the use of GFAP; however, conclusive results have not been obtained, which is also attributed to analytical heterogeneity among laboratories, so a direct comparison across studies was unavailable.
Additionally, in experimental rodent models of both cerebrovascular and traumatic brain injury, S100B appears to actively participate in the pathogenic processes. In both experimental animal models (middle cerebral artery occlusion (MCAO) and controlled cortical impact), high levels of S100B were detected in injured tissues [76,77,78]. Inherently, larger infarct volumes and worse neurological deficits after permanent MCAO were shown in mice overexpressing S100B (TghuS100B mice) as compared to wild-type mice [79]. Likewise, in MCAO mice, S100B treatment inhibited M2 polarization, which is regarded to be anti-inflammatory and neuroprotective, while promoting microglia M1 polarization, which is regarded to induce inflammation and neurotoxicity, with enhanced migration ability and aggravated cerebral ischemia [80]. On the contrary, the pharmacological AA-dependent inhibition of astrocytic S100B synthesis was accompanied by the prevention of brain damage and neurological deficits, as well as delayed infarct expansion, even reducing neuroinflammatory processes and motor deficits in experimental animals with intracerebral hemorrhage or subdural hematomas [81,82,83,84]. These observations confirmed a putative role played by S100B in acute brain injury pathogenic processes. Likewise, treatment with a neutralizing anti-S100B antibody in mice subjected to controlled cortical impact attenuated microglial activation, reduced lesion size, improved neuronal survival, and induced significant improvements in sensorimotor performance and memory retention as compared with control mice treated with normal IgG or vehicle [85]. In addition, the AA-induced inhibition of S100B synthesis has been shown to recover tissue damage and memory deficits produced by hypoxia-ischemia in neonatal rats [86], while in rats prone to spontaneously hypertensive stroke, AA prevented hypertension-induced stroke and inhibited the enlargement of the stroke lesion [87]. Additionally, in rats with spinal cord injury, the AA-dependent inhibition of S100B synthesis was accompanied by a reduction of secondary lesions, an improvement of motor function [88], and suppressed neuropathic pain [89]. In addition, in S100B knockout mice, where the protein was not synthesized, functional and neuropathological ameliorations were observed after traumatic brain injury [85], and similar results were observed in rats with ischemic stroke silenced for S100B using RNA interference [90].
Interestingly, it should also be noted, on the other hand, that increased dentate neurogenesis [91], as well as hippocampal synaptogenesis [92], have been reported after intraventricular administration of S100B in an experimental model of traumatic brain injury. This apparent discrepancy might be attributed to the different roles (trophic or toxic) played by the protein at different concentrations.

1.6. S100B in Epilepsy

Although the origin of epileptic seizures is known to be heterogeneous, many studies have been performed to detect biomarkers that can aid the diagnosis and therapy of different forms of epileptic seizures, aiming at the identification of subjects at risk of epilepsy development and monitoring the disease [93,94,95]. Among these studies, high levels of S100B protein in adults and children with different forms of epileptic seizures have been described [96,97,98,99,100,101,102,103,104,105,106,107]. In addition, it has been reported that anti-seizure therapies such as carbamazepine, oxcarbazepine, or levetiracetam were able to significantly reduce serum levels of S100B [108,109,110].
The potential involvement of S100B in the etiopathology of epilepsy has been investigated in distinct animal models. The administration to adult rats of kainic acid, which triggers seizures and neuronal loss in a manner that mirrors the neuropathology of human epilepsy, was shown to increase S100B expression in the hippocampus as a marker of activation of a definite subpopulation of astrocytes [111] and as an inflammatory cytokine [112]. Conversely, treatment with Minozac, an inhibitor of proinflammatory cytokine upregulation, was accompanied by a reduction of S100B and other indicators of glial activation, also inducing behavioral improvement [113]. Inherently, an anti-inflammatory drug such as metformin was shown to reduce S100B brain levels in a kainic-acid-induced model of epilepsy [113]. Likewise, levels of S100B secreted from hippocampal slices from a pilocarpine-induced model of epilepsy were reduced by dexamethasone treatment [114]. Similarly, rats with chronic epilepsy, when treated with the anti-epileptic agent resveratrol, had reduced S100B levels in both CSF and blood [115]. In accordance with the above-described results, also in this disorder, as in other above-mentioned diseases, the inhibitor of astrocytic S100B synthesis, AA, has been shown to produce clinical amelioration. In particular, the downregulation of neuroinflammatory parameters and astrocyte dysfunction in young rats after status epilepticus induced by Li-pilocarpine were observed [116]. In summary, studies addressing S100B as a biomarker and/or pathogenic factor of epileptic seizures, possibly as a consequence of neuroinflammatory processes proper to these disorders, have constituted an active research trend in recent years.
Intriguingly, S100B knockout mice lacking a functional S100B gene, thus not expressing the S100B protein, have been reported to be subject to earlier and more severe seizures than wild-type mice [117]. Indeed, these results do not appear to fit the bulk of the above-reported data. Peculiarities of the epileptogenic procedure used (electrical kindling of the amygdala) or unknown consequences accompanying the S100B knockout procedure might explain these apparent discrepancies.

1.7. S100B in Inflammatory Bowel Disease

Converging evidence indicates a possible involvement of the S100B protein in the pathogenic processes of inflammatory bowel disease (IBD), a term indicating two conditions (Crohn’s disease and ulcerative colitis) that are characterized by chronic inflammation of the gastrointestinal tract. The enteric nervous system, putatively interacting with the gut microbiota, a population of microorganisms that influences the immune system of the host during homeostasis and disease, actively participates in IBD processes [118,119,120,121,122]. S100B is present and plays a key role in enteroglial cells, which resemble astrocytes of the central nervous system, as well as in inflammatory processes [123,124]. Taken together, these data suggest the possibility that S100B may be involved in IBD pathogenic processes [125].
Indeed, considering S100B as a possible IBD biomarker in biological fluids, its levels were found to be lower in patients than in healthy subjects both in serum [126] and in feces [127], while the levels of S100B in biological fluids were found to be higher in all the pathological conditions investigated as compared with healthy subjects. Possible interactions with the microbiota and/or structural/functional peculiarities of the gastroenteric compartment might explain this discrepancy.
In any case, S100B in human enteroglial cells has been shown to be overexpressed in subjects affected by IBD, to stimulate NO production, and to correlate with the gut’s inflammatory status [128]. Similarly, in the animal model of acute colitis induced by dextran sodium sulfate, high levels of S100B were described, while macroscopic and histological/biochemical assays of colonic tissues and plasma revealed a significant amelioration after treatment with PTM, the inhibitor of S100B activity [129]. PTM also prevented intestinal inflammation, oxidative stress, enteric glia activation, neuronal loss, and histological injury in intestinal mucositis induced by 5-fluorouracil, blocking an S100B-RAGE-NFkB pathway [130]. Noteworthy, a common pathway involving S100B up-regulation and Toll-like receptor (a receptor recognizing DAMPs such as S100B) stimulation has been described in the inflamed colon [131,132]. Additionally, enteric glia-derived S100B has been proposed as a putative bridge linking colonic inflammation and cancer of the colon, given its ability to interact with factors involved in both conditions (NF-κB, RAGE, and p53). Consequently, the S100B inhibitor PTM has also been proposed as a putative anti-cancer drug [133].
Finally, as above indicated, based on the consideration that a decrease in the abundance and diversity of gut microbiota-specific genera may putatively trigger IBD-initiating events [134], the possible interactions of S100B with microbiota have been recently investigated both in silico [135] and experimentally in vivo in mice [136]. Microbiota proteins putatively interacting with S100B domains were found to be reduced in IBD patients with respect to healthy subjects, also exhibiting differences in the occurrence of interacting domains. Interestingly, these in silico inferences were experimentally confirmed in mice. In fact, S100B levels were experimentally shown to correlate with microbiota biodiversity, and the correlation was significantly reduced after treatment with the S100B inhibitor PTM [136]. These data may open novel perspectives about the potential role of S100B in the gut both as a constituent of enteroglial glial cells [123], which may release it, and putatively as a constituent of food such as milk [137,138]. Thus, the protein might mediate the regulation in the intestinal microbiota, also potentially influencing IBD pathogenic processes.

1.8. Perspectives

Collectively, the above data delineate a scenario where the S100B protein stands at the crossroads of different pathological conditions. Its pathogenic role has been proposed in processes caused by different etiologic factors and displaying different symptoms [3,5]. However, these disorders, regardless of their origin, appear to involve processes that share aspects attributable to inflammation [139]. This view might depict S100B as an unspecific putative pathogenic factor, comprehensibly regarded with some suspicion. However, this view also presents S100B as a wide-ranging tool that may reasonably lead to unifying solutions, thus offering a promising perspective. Interestingly in this respect, the range of diseases where S100B is putatively involved in pathogenic processes, at least based on results in experimental models, is now enlarging outside the nervous system (e.g., muscular dystrophy, obesity, diabetes, ocular disorders); potentially, it is expected to parallel the cellular distribution of the protein, which is definite but significant [3,5]. Indeed, at present, the S100B protein is not the unique inflammatory molecule for which a wide therapeutic target role is putatively recognized. As examples, the prototype of DAMP proteins, HMGB1, as well as the inflammatory mediators’ resistin-like molecules, are currently regarded as interesting unifying therapeutic targets for different disorders [140,141,142]. It should also be noted in this respect that RAGE ligands, including DAMPs, have been considered potential therapeutic targets for a variety of disorders [143].
Thus, drugs able to efficaciously counteract S100B have been actively searched and individuated and are currently searched. Some molecules, such as AA [82], PTM [144], and TRTK12 peptide [145], have been individuated in animal models, cell cultures, or even by screening a bacteriophage random peptide display library, respectively. For PTM, an intranasal delivery system aimed at more easily reaching the nervous system protected by the blood-–brain barrier (chitosan coated niosomes) has also been proposed [133]. While the mechanism by which AA inhibits the synthesis of astrocytic S100B is still unclear, PTM and TRTK12 are known to block the interaction between S100B and the transcription factor p53. In any case, the use of AA and PTM has been widely validated in animal experimental models of different diseases, as indicated above. It is also relevant that various approaches and investigations to individuate additional, even more efficacious, molecules able to counteract S100B are currently in progress, mostly using stereological procedures [145,146,147,148]. A necessary step towards a clinical setting for S100B antagonists will be clinical trials. Clinical trials on the use of AA in neurodegenerative disorders (Alzheimer’s and Parkinson’s diseases, amyotrophic lateral sclerosis) have already been proposed and completed, although results do not appear to be available (NCT00694941, NCT00212693, NCT00403104, NCT00083421).
In addition, owing to the recognized role of S100B as a biomarker, the clinical targeting of S100B at present may reasonably be supported by the standard hospital-based tests that measure this molecule in biological fluids. Mostly, these tests are based on the widely diffused ELISA method, and have already been tuned; numerous kits for measuring S100B in human biological fluids are commercially available. This multifaceted but unifying view of S100B as a pathogenic factor may reasonably stimulate research aimed at clarifying putatively common pathogenic processes in a variety of disorders, apparently heterogeneous for symptoms, target districts, and etiologies, but sharing the dysregulation of a multifaceted pathogenic factor as the S100B protein.

Funding

We are grateful to the Nando and Elsa Peretti Foundation for support.

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. Moore, B.W. A soluble protein characteristic of the nervous system. Biochem. Biophys. Res. Commun. 1965, 19, 739–744. [Google Scholar] [CrossRef] [PubMed]
  2. Grzybowska, E.A. Calcium-binding proteins with disordered structure and their role in secretion, storage, and cellular signaling. Biomolecules 2018, 8, 42. [Google Scholar] [CrossRef]
  3. Michetti, F.; D’Ambrosi, N.; Toesca, A.; Puglisi, M.A.; Serrano, A.; Marchese, E.; Corvino, V.; Geloso, M.C. The S100B story: From biomarker to active factor in the neural injury. J. Neurochem. 2019, 148, 168–187. [Google Scholar] [CrossRef]
  4. Goswami, D.; Anuradha, U.; Angati, A.; Kumari, N.; Singh, R.K. Pharmacological and pathological relevance of S100 proteins in neurological disorders. CNS Neurol. Disord. Drug Targets 2022, 22. Online ahead of print. [Google Scholar]
  5. Michetti, F.; Di Sante, G.; Clementi, M.E.; Sampaolese, B.; Casalbore, P.; Volonté, C.; Romano Spica, V.; Parnigotto, P.P.; Di Liddo, R.; Amadio, S.; et al. Growing role of S100B protein as a putative therapeutic target for neurological- and nonneurological-disorders. Neurosci. Biobehav. Rev. 2021, 127, 446–458. [Google Scholar] [CrossRef] [PubMed]
  6. Langeh, U.; Singh, S. Targeting S100B Protein as a Surrogate Biomarker and its Role in Various Neurological Disorders. Curr. Neuropharmacol. 2021, 19, 265–277. [Google Scholar] [CrossRef] [PubMed]
  7. Zaręba-Kozioł, M.; Burdukiewicz, M.; Wysłouch-Cieszyńska, A. Intracellular Protein S-Nitrosylation-A Cells Response to Extracellular S100B and RAGE Receptor. Biomolecules 2022, 12, 613. [Google Scholar] [CrossRef]
  8. Michetti, F.; Corvino, V.; Geloso, M.C.; Lattanzi, W.; Bernardini, C.; Serpero, L.; Gazzolo, D. The S100B protein in biological fluids: More than a lifelong biomarker of brain distress. J. Neurochem. 2012, 120, 644–659. [Google Scholar] [CrossRef]
  9. Janigro, D.; Mondello, S.; Posti, J.P.; Unden, J. GFAP and S100B: What you always wanted to know and never dared to ask. Front. Neurol. 2022, 13, 835597. [Google Scholar] [CrossRef]
  10. Aceti, A.; Margarucci, L.M.; Scaramucci, E.; Orsini, M.; Salerno, G.; Di Sante, G.; Gianfranceschi, G.; Di Liddo, R.; Valeriani, F.; Ria, F.; et al. Serum S100B protein as a marker of severity in Covid-19 patients. Sci. Rep. 2020, 10, 18665. [Google Scholar] [CrossRef]
  11. Rodríguez-Giraldo, M.; González-Reyes, R.E.; Ramírez-Guerrero, S.; Bonilla-Trilleras, C.E.; Guardo-Maya, S.; Nava-Mesa, M.O. Astrocytes as a Therapeutic Target in Alzheimer’s Disease-Comprehensive Review and Recent Developments. Int. J. Mol. Sci. 2022, 23, 13630. [Google Scholar] [CrossRef]
  12. Mori, T.; Town, T.; Tan, J.; Yada, N.; Horikoshi, Y.; Yamamoto, J.; Shimoda, T.; Kamanaka, Y.; Tateishi, N.; Asano, T. Arundic Acid ameliorates cerebral amyloidosis and gliosis in Alzheimer transgenic mice. J. Pharmacol. Exp. Ther. 2006, 318, 571–578. [Google Scholar] [CrossRef] [PubMed]
  13. Griffin, W.S.; Stanley, L.C.; Ling, C.; White, L.; MacLeod, V.; Perrot, L.J.; White, C.L., 3rd; Araoz, C. Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc. Natl. Acad. Sci. USA 1989, 86, 7611–7615. [Google Scholar] [CrossRef]
  14. Marshak, D.R.; Pesce, S.A.; Stanley, L.C.; Griffin, W.S. Increased S100 beta neurotrophic activity in Alzheimer’s disease temporal lobe. Neurobiol Aging. 1992, 13, 1–7. [Google Scholar] [CrossRef] [PubMed]
  15. Van Eldik, L.J.; Griffin, W.S. S100 beta expression in Alzheimer’s disease: Relation to neuropathology in brain regions. Biochim. Biophys. Acta 1994, 1223, 398–403. [Google Scholar] [CrossRef] [PubMed]
  16. Mrak, R.E.; Sheng, J.G.; Griffin, W.S. Correlation of astrocytic S100 beta expression with dystrophic neurites in amyloid plaques of Alzheimer’s disease. J. Neuropathol. Exp. Neurol. 1996, 55, 273–279. [Google Scholar] [CrossRef]
  17. Bellaver, B.; Ferrari-Souza, J.P.; Uglione da Ros, L.; Carter, S.F.; Rodriguez-Vieitez, E.; Nordberg, A.; Pellerin, L.; Rosa-Neto, P.; Leffa, D.T.; Zimmer, E.R. Astrocyte Biomarkers in Alzheimer Disease: A Systematic Review and Meta-analysis. Neurology 2021, 96, e2944–e2955. [Google Scholar] [CrossRef]
  18. Salvadó, G.; Shekari, M.; Falcon, C.; Operto, G.; Milà-Alomà, M.; Sánchez-Benavides, G.; Cacciaglia, R.; Arenaza-Urquijo, E.; Niñerola-Baizán, A.; Perissinotti, A.; et al. Brain alterations in the early Alzheimer’s continuum with amyloid-β, tau, glial and neurodegeneration CSF markers. Brain Commun. 2022, 4, fcac134. [Google Scholar] [CrossRef]
  19. Mori, T.; Koyama, N.; Arendash, G.W.; Horikoshi-Sakuraba, Y.; Tan, J.; Town, T. Overexpression of human S100B exacerbates cerebral amyloidosis and gliosis in the Tg2576 mouse model of Alzheimer’s disease. Glia 2010, 58, 300–314. [Google Scholar] [CrossRef]
  20. Wartchow, K.M.; Rodrigues, L.; Swierzy, I.; Buchfelder, M.; de Souza, D.O.; Gonçalves, C.A.; Kleindienst, A. Amyloid-β Processing in Aged S100B Transgenic Mice Is Sex Dependent. Int. J. Mol. Sci. 2021, 22, 10823. [Google Scholar] [CrossRef]
  21. Cirillo, C.; Capoccia, E.; Iuvone, T.; Cuomo, R.; Sarnelli, G.; Steardo, L.; Esposito, G. S100B Inhibitor Pentamidine Attenuates Reactive Gliosis and Reduces Neuronal Loss in a Mouse Model of Alzheimer’s Disease. Biomed. Res. Int. 2015, 2015, 508342. [Google Scholar] [CrossRef]
  22. Asano, T.; Mori, T.; Shimoda, T.; Shinagawa, R.; Satoh, S.; Yada, N.; Katsumata, S.; Matsuda, S.; Kagamiishi, Y.; Tateishi, N. Arundic acid (ONO-2506) ameliorates delayed ischemic brain damage by preventing astrocytic overproduction of S100B. Curr. Drug Target-CNS Neurol. Disord. 2005, 4, 127–142. [Google Scholar] [CrossRef]
  23. Roltsch, E.; Holcomb, L.; Young, K.A.; Marks, A.; Zimmer, D.B. PSAPP mice exhibit regionally selective reductions in gliosis and plaque deposition in response to S100B ablation. J. Neuroinflamm. 2010, 7, 78. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, J.; Zhou, Y.; Yang, Y.; Gao, X.; Liu, Z.; Hong, G.; Yao, L.; Yin, J.; Gu, X.; Li, K. S100B gene polymorphisms are associated with the S100B level and Alzheimer’s disease risk by altering the miRNA binding capacity. Aging 2021, 13, 13954–13967. [Google Scholar] [CrossRef] [PubMed]
  25. Figueira, A.J.; Saavedra, J.; Cardoso, I.; Gomes, C.M. S100B chaperone multimers suppress the formation of oligomers during Aβ42 aggregation. Front. Neurosci. 2023, 17, 1162741. [Google Scholar] [CrossRef] [PubMed]
  26. Rydbirk, R.; Elfving, B.; Andersen, M.D.; Langbøl, M.A.; Folke, J.; Winge, K.; Pakkenberg, B.; Brudek, T.; Aznar, S. Cytokine profiling in the prefrontal cortex of Parkinson’s Disease and Multiple System Atrophy patients. Neurobiol. Dis. 2017, 106, 269–278. [Google Scholar] [CrossRef]
  27. Sathe, K.; Maetzler, W.; Lang, J.D.; Mounsey, R.B.; Fleckenstein, C.; Martin, H.L.; Schulte, C.; Mustafa, S.; Synofzik, M.; Vukovic, Z.; et al. S100B is increased in Parkinson’s disease and ablation protects against MPTP-induced toxicity through the RAGE and TNF-α pathway. Brain 2012, 135 Pt 11, 3336–3347. [Google Scholar] [CrossRef]
  28. Viana, S.D.; Valero, J.; Rodrigues-Santos, P.; Couceiro, P.; Silva, A.M.; Carvalho, F.; Ali, S.F.; Fontes-Ribeiro, C.A.; Pereira, F.C. Regulation of striatal astrocytic receptor for advanced glycation end-products variants in an early stage of experimental Parkinson’s disease. J. Neurochem. 2016, 138, 598–609. [Google Scholar] [CrossRef]
  29. Liu, J.; Wang, H.; Zhang, L.; Xu, Y.; Deng, W.; Zhu, H.; Qin, C. S100B transgenic mice develop features of Parkinson’s disease. Arch. Med. Res. 2011, 42, 1–7. [Google Scholar] [CrossRef]
  30. Bancroft, E.A.; De La Mora, M.; Pandey, G.; Zarate, S.M.; Srinivasan, R. Extracellular S100B inhibits A-type voltage-gated potassium currents and increases L-type voltage-gated calcium channel activity in dopaminergic neurons. Glia 2022, 70, 2330–2347. [Google Scholar] [CrossRef] [PubMed]
  31. Carvalho, D.Z.; Schönwald, S.V.; Schumacher-Schuh, A.F.; Braga, C.W.; Souza, D.O.; Oses, J.P.; Donis, K.C.; Rieder, C.R. Overnight S100B in Parkinson’s Disease: A glimpse into sleep-related neuroinflammation. Neurosci. Lett. 2015, 608, 57–63. [Google Scholar] [CrossRef] [PubMed]
  32. Fardell, C.; Zettergren, A.; Ran, C.; Carmine Belin, A.; Ekman, A.; Sydow, O.; Bäckman, L.; Holmberg, B.; Dizdar, N.; Söderkvist, P.; et al. S100B polymorphisms are associated with age of onset of Parkinson’s disease. BMC Med. Genet. 2018, 19, 42. [Google Scholar] [CrossRef] [PubMed]
  33. Angelopoulou, E.; Paudel, Y.N.; Piperi, C. Emerging role of S100B protein implication in Parkinson’s disease pathogenesis. Cell. Mol. Life Sci. 2021, 78, 1445–1453. [Google Scholar] [CrossRef]
  34. Rinaldi, F.; Seguella, L.; Gigli, S.; Hanieh, P.N.; Del Favero, E.; Cantù, L.; Pesce, M.; Sarnelli, G.; Marianecci, C.; Esposito, G.; et al. inPentasomes: An innovative nose-to-brain pentamidine delivery blunts MPTP parkinsonism in mice. J. Control. Release 2019, 294, 17–26. [Google Scholar] [CrossRef]
  35. Kato, H.; Kurosaki, R.; Oki, C.; Araki, T. Arundic acid, an astrocyte-modulating agent, protects dopaminergic neurons against MPTP neurotoxicity in mice. Brain Res. 2004, 1030, 66–73. [Google Scholar] [CrossRef]
  36. Kamo, H.; Haebara, H.; Akiguchi, I.; Kameyama, M.; Kimura, H.; McGeer, P.L. A distinctive distribution of reactive astroglia in the precentral cortex in amyotrophic lateral sclerosis. Acta Neuropathol. 1987, 74, 33–38. [Google Scholar] [CrossRef] [PubMed]
  37. Migheli, A.; Cordera, S.; Bendotti, C.; Atzori, C.; Piva, R.; Schiffer, D. S-100beta protein is upregulated in astrocytes and motor neurons in the spinal cord of patients with amyotrophic lateral sclerosis. Neurosci. Lett. 1999, 261, 25–28. [Google Scholar] [CrossRef] [PubMed]
  38. Otto, M.; Bahn, E.; Wiltfang, J.; Boekhoff, I.; Beuche, W. Decrease of S100 beta protein in serum of patients with amyotrophic lateral sclerosis. Neurosci. Lett. 1998, 240, 171–173. [Google Scholar] [CrossRef] [PubMed]
  39. Süssmuth, S.D.; Sperfeld, A.D.; Hinz, A.; Brettschneider, J.; Endruhn, S.; Ludolph, A.C.; Tumani, H. CSF glial markers correlate with survival in amyotrophic lateral sclerosis. Neurology 2010, 74, 982–987. [Google Scholar] [CrossRef]
  40. Steinacker, P.; Huss, A.; Mayer, B.; Grehl, T.; Grosskreutz, J.; Borck, G.; Kuhle, J.; Lulé, D.; Meyer, T.; Oeckl, P.; et al. Diagnostic and prognostic significance of neurofilament light chain NF-L, but not progranulin and S100B, in the course of amyotrophic lateral sclerosis: Data from the German MND-net. Amyotroph. Lateral Scler. Front. Degener. 2017, 18, 112–119. [Google Scholar] [CrossRef]
  41. Nowicka, N.; Szymańska, K.; Juranek, J.; Zglejc-Waszak, K.; Korytko, A.; Załęcki, M.; Chmielewska-Krzesińska, M.; Wąsowicz, K.; Wojtkiewicz, J. The Involvement of RAGE and Its Ligands during Progression of ALS in SOD1 G93A Transgenic Mice. Int. J. Mol. Sci. 2022, 23, 2184. [Google Scholar] [CrossRef] [PubMed]
  42. Díaz-Amarilla, P.; Olivera-Bravo, S.; Trias, E.; Cragnolini, A.; Martínez-Palma, L.; Cassina, P.; Beckman, J.; Barbeito, L. Phenotypically aberrant astrocytes that promote motoneuron damage in a model of inherited amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA 2011, 108, 18126–18131. [Google Scholar] [CrossRef] [PubMed]
  43. Serrano, A.; Donno, C.; Giannetti, S.; Perić, M.; Andjus, P.; D’Ambrosi, N.; Michetti, F. The Astrocytic S100B Protein with Its Receptor RAGE Is Aberrantly Expressed in SOD1G93A Models, and Its Inhibition Decreases the Expression of Proinflammatory Genes. Mediators Inflamm. 2017, 2017, 1626204. [Google Scholar] [CrossRef] [PubMed]
  44. Michetti, F.; Massaro, A.; Murazio, M. The nervous system-specific S-100 antigen in cerebrospinal fluid of multiple sclerosis patients. Neurosci. Lett. 1979, 11, 171–175. [Google Scholar] [CrossRef]
  45. Barateiro, A.; Afonso, V.; Santos, G.; Cerqueira, J.J.; Brites, D.; van Horssen, J.; Fernandes, A. S100B as a Potential Biomarker and Therapeutic Target in Multiple Sclerosis. Mol. Neurobiol. 2016, 53, 3976–3991. [Google Scholar] [CrossRef]
  46. Petzold, A.; Eikelenboom, M.J.; Gveric, D.; Keir, G.; Chapman, M.; Lazeron, R.H.; Cuzner, M.L.; Polman, C.H.; Uitdehaag, B.M.; Thompson, E.J.; et al. Markers for different glial cell responses in multiple sclerosis: Clinical and pathological correlations. Brain 2002, 125 Pt 7, 1462–1473. [Google Scholar] [CrossRef]
  47. Yan, S.S.; Wu, Z.Y.; Zhang, H.P.; Furtado, G.; Chen, X.; Yan, S.F.; Schmidt, A.M.; Brown, C.; Stern, A.; LaFaille, J.; et al. Suppression of experimental autoimmune encephalomyelitis by selective blockade of encephalitogenic T-cell infiltration of the central nervous system. Nat. Med. 2003, 9, 287–293. [Google Scholar] [CrossRef]
  48. Santos, G.; Barateiro, A.; Gomes, C.M.; Brites, D.; Fernandes, A. Impaired oligodendrogenesis and myelination by elevated S100B levels during neurodevelopment. Neuropharmacology 2018, 129, 69–83. [Google Scholar] [CrossRef]
  49. Di Sante, G.; Amadio, S.; Sampaolese, B.; Clementi, M.E.; Valentini, M.; Volonté, C.; Casalbore, P.; Ria, F.; Michetti, F. The S100B Inhibitor Pentamidine Ameliorates Clinical Score and Neuropathology of Relapsing-Remitting Multiple Sclerosis Mouse Model. Cells 2020, 9, 748. [Google Scholar] [CrossRef]
  50. Camponeschi, C.; De Carluccio, M.; Amadio, S.; Clementi, M.E.; Sampaolese, B.; Volonté, C.; Tredicine, M.; Romano Spica, V.; Di Liddo, R.; Ria, F.; et al. S100B Protein as a Therapeutic Target in Multiple Sclerosis: The S100B Inhibitor Arundic Acid Protects from Chronic Experimental Autoimmune Encephalomyelitis. Int. J. Mol. Sci. 2021, 22, 13558. [Google Scholar] [CrossRef]
  51. Barros, C.; Barateiro, A.; Neto, A.; Soromenho, B.; Basto, A.P.; Mateus, J.M.; Xapelli, S.; Sebastião, A.M.; Brites, D.; Graça, L.; et al. S100B inhibition protects from chronic experimental autoimmune encephalomyelitis. Brain Commun. 2022, 4, fcac076. [Google Scholar] [CrossRef]
  52. Minkkinen, M.; Iverson, G.L.; Kotilainen, A.K.; Pauniaho, S.L.; Mattila, V.M.; Lehtimäki, T.; Berghem, K.; Posti, J.P.; Luoto, T.M. Prospective Validation of the Scandinavian Guidelines for Initial Management of Minimal, Mild, and Moderate Head Injuries in Adults. J. Neurotrauma 2019, 36, 2904–2912. [Google Scholar] [CrossRef]
  53. Rogan, A.; Sik, A.; Dickinson, E.; Patel, V.; Peckler, B.; McQuade, D.; Larsen, P.D.; Endorsed by ACEM Emergency Department Epidemiology Network. Diagnostic performance of S100B as a rule-out test for intracranial pathology in head-injured patients presenting to the emergency department who meet NICE Head Injury Guideline criteria for CT-head scan. Emerg. Med. J. 2023, 40, 159–166. [Google Scholar] [CrossRef]
  54. Rossi, R.; Douglas, A.; Gil, S.M.; Jabrah, D.; Pandit, A.; Gilvarry, M.; McCarthy, R.; Prendergast, J.; Jood, K.; Redfors, P.; et al. S100b in acute ischemic stroke clots is a biomarker for post-thrombectomy intracranial hemorrhages. Front. Neurol. 2023, 13, 1067215. [Google Scholar] [CrossRef] [PubMed]
  55. Glimmerveen, A.; Verhulst, M.; Verbunt, J.; Van Heugten, C.; Hofmeijer, J. Predicting Long-Term Cognitive Impairments in Survivors after Cardiac Arrest: A Systematic Review. J. Rehabil. Med. 2023, 55, jrm00368. [Google Scholar] [CrossRef] [PubMed]
  56. Si, Y.; Duan, W.; Xie, J.; Duan, C.; Liu, S.; Wang, Q.; Zhao, X.; Wu, D.; Wang, Y.; Wang, L.; et al. Biomarkers for prediction of neurological complications after acute Stanford type A aortic dissection: A systematic review and meta-analysis. PLoS ONE 2023, 18, e0281352. [Google Scholar] [CrossRef] [PubMed]
  57. Dzierzęcki, S.; Ząbek, M.; Zapolska, G.; Tomasiuk, R. The S-100B level, intracranial pressure, body temperature, and transcranial blood flow velocities predict the outcome of the treatment of severe brain injury. Medicine 2022, 101, e30348. [Google Scholar] [CrossRef]
  58. Seidenfaden, S.C.; Kjerulff, J.L.; Juul, N.; Kirkegaard, H.; Fogh Møller, M.; Bloch Münster, A.M.; Thingemann Bøtker, M. Temporal Changes in Serum S100B Levels from Prehospital to Early In-Hospital Sampling in Patients Suffering Traumatic Brain Injury. Front. Neurol. 2022, 13, 800015. [Google Scholar] [CrossRef]
  59. Gradisek, P.; Carrara, G.; Antiga, L.; Bottazzi, B.; Chieregato, A.; Csomos, A.; Fainardi, E.; Filekovic, S.; Fleming, J.; Hadjisavvas, A.; et al. Prognostic Value of a Combination of Circulating Biomarkers in Critically Ill Patients with Traumatic Brain Injury: Results from the European CREACTIVE Study. J. Neurotrauma 2021, 38, 2667–2676. [Google Scholar] [CrossRef]
  60. Lin, I.H.; Kamnaksh, A.; Aniceto, R.; McCullough, J.; Bekdash, R.; Eklund, M.; Ghatan, P.H.; Risling, M.; Svensson, M.; Bellander, B.M.; et al. Time-Dependent Changes in the Biofluid Levels of Neural Injury Markers in Severe Traumatic Brain Injury Patients-Cerebrospinal Fluid and Cerebral Microdialysates: A Longitudinal Prospective Pilot Study. Neurotrauma Rep. 2023, 4, 107–117. [Google Scholar] [CrossRef]
  61. Rogan, A.; O’Sullivan, M.B.; Holley, A.; McQuade, D.; Larsen, P. Can serum biomarkers be used to rule out significant intracranial pathology in emergency department patients with mild traumatic brain injury? A Systemic Review & Meta-Analysis. Injury 2022, 53, 259–271. [Google Scholar]
  62. Sapin, V.; Gaulmin, R.; Aubin, R.; Walrand, S.; Coste, A.; Abbot, M. Blood biomarkers of mild traumatic brain injury: State of art. Neurochirurgie 2021, 67, 249–254. [Google Scholar] [CrossRef] [PubMed]
  63. Amoo, M.; Henry, J.; O’Halloran, P.J.; Brennan, P.; Husien, M.B.; Campbell, M.; Caird, J.; Javadpour, M.; Curley, G.F. S100B, GFAP, UCH-L1 and NSE as predictors of abnormalities on CT imaging following mild traumatic brain injury: A systematic review and meta-analysis of diagnostic test accuracy. Neurosurg. Rev. 2022, 45, 1171–1193. [Google Scholar] [CrossRef]
  64. Faisal, M.; Vedin, T.; Edelhamre, M.; Forberg, J.L. Diagnostic performance of biomarker S100B and guideline adherence in routine care of mild head trauma. Scand. J. Trauma Resusc. Emerg. Med. 2023, 31, 3. [Google Scholar] [CrossRef] [PubMed]
  65. Hopman, J.H.; Santing, J.A.L.; Foks, K.A.; Verheul, R.J.; van der Linden, C.M.; van den Brand, C.L.; Jellema, K. Biomarker S100B in plasma a screening tool for mild traumatic brain injury in an emergency department. Brain Inj. 2023, 37, 47–53. [Google Scholar] [CrossRef]
  66. Chen, H.; Ding, V.Y.; Zhu, G.; Jiang, B.; Li, Y.; Boothroyd, D.; Rezaii, P.G.; Bet, A.M.; Paulino, A.D.; Weber, A.; et al. Association between Blood and Computed Tomographic Imaging Biomarkers in a Cohort of Mild Traumatic Brain Injury Patients. J. Neurotrauma 2022, 39, 1329–1338. [Google Scholar] [CrossRef] [PubMed]
  67. Meshkini, A.; Ghorbani Haghjo, A.; Hasanpour Segherlou, Z.; Nouri-Vaskeh, M. S100 Calcium-Binding Protein B and Glial Fibrillary Acidic Protein in Patients with Mild Traumatic Brain Injury. Bull. Emerg. Trauma 2021, 9, 183–187. [Google Scholar]
  68. Mastandrea, P.; Mengozzi, S.; Bernardini, S. Systematic review and cumulative meta-analysis of the diagnostic accuracy of glial fibrillary acidic protein vs. S100 calcium binding protein B as blood biomarkers in observational studies of patients with mild or moderate acute traumatic brain injury. Diagnosis 2021, 9, 18–27. [Google Scholar] [CrossRef]
  69. Seidenfaden, S.C.; Kjerulff, J.L.; Juul, N.; Kirkegaard, H.; Møller, M.F.; Münster, A.B.; Bøtker, M.T. Diagnostic accuracy of prehospital serum S100B and GFAP in patients with mild traumatic brain injury: A prospective observational multicenter cohort study—“The PreTBI I study”. Scand. J. Trauma Resusc. Emerg. Med. 2021, 29, 75. [Google Scholar] [CrossRef]
  70. Oris, C.; Bouillon-Minois, J.B.; Pinguet, J.; Kahouadji, S.; Durif, J.; Meslé, V.; Pereira, B.; Schmidt, J.; Sapin, V.; Bouvier, D. Predictive Performance of Blood S100B in the Management of Patients Over 65 Years Old with Mild Traumatic Brain Injury. J. Gerontol. A Biol. Sci. Med. Sci. 2021, 76, 1471–1479. [Google Scholar] [CrossRef]
  71. Oris, C.; Kahouadji, S.; Durif, J.; Bouvier, D.; Sapin, V. S100B, Actor and Biomarker of Mild Traumatic Brain Injury. Int. J. Mol. Sci. 2023, 24, 6602. [Google Scholar] [CrossRef] [PubMed]
  72. Kleindienst, A.; Ross Bullock, M. A critical analysis of the role of the neurotrophic protein S100B in acute brain injury. J. Neurotrauma 2006, 23, 1185–1200. [Google Scholar] [CrossRef] [PubMed]
  73. Müller, M.; Münster, J.M.; Hautz, W.E.; Gerber, J.L.; Schefold, J.C.; Exadaktylos, A.K.; Pfortmueller, C.A. Increased S-100 B levels are associated with fractures and soft tissue injury in multiple trauma patients. Injury 2020, 51, 812–818. [Google Scholar] [CrossRef] [PubMed]
  74. Hier, D.B.; Obafemi-Ajayi, T.; Thimgan, M.S.; Olbricht, G.R.; Azizi, S.; Allen, B.; Hadi, B.A.; Wunsch, D.C., 2nd. Blood biomarkers for mild traumatic brain injury: A selective review of unresolved issues. Biomark. Res. 2021, 9, 70. [Google Scholar] [CrossRef]
  75. Grau-Mercier, L.; Grandpierre, R.G.; Alonso, S.; Savey, A.; Le Floch, A.; de Oliveira, F.; Masia, T.; Jory, N.; Coisy, F.; Claret, P.G. S100B serum level: A relevant biomarker for the management of non-traumatic headaches in emergency care? Am. J. Emerg. Med. 2023, 68, 132–137. [Google Scholar] [CrossRef]
  76. Matsui, T.; Mori, T.; Tateishi, N.; Kagamiishi, Y.; Satoh, S.; Katsube, N.; Morikawa, E.; Morimoto, T.; Ikuta, F.; Asano, T. Astrocytic activation and delayed infarct expansion after permanent focal ischemia in rats. Part I: Enhanced astrocytic synthesis of s-100beta in the periinfarct area precedes delayed infarct expansion. J. Cereb. Blood Flow Metab. 2002, 22, 711–722. [Google Scholar] [CrossRef]
  77. Kleindienst, A.; Tolias, C.M.; Corwin, F.D.; Müller, C.; Marmarou, A.; Fatouros, P.; Bullock, M.R. Assessment of cerebral S100B levels by proton magnetic resonance spectroscopy after lateral fluid-percussion injury in the rat. J. Neurosurg. 2005, 102, 1115–1121. [Google Scholar] [CrossRef]
  78. Sandhir, R.; Onyszchuk, G.; Berman, N.E. Exacerbated glial response in the aged mouse hippocampus following controlled cortical impact injury. Exp. Neurol. 2008, 213, 372–380. [Google Scholar] [CrossRef]
  79. Mori, T.; Tan, J.; Arendash, G.W.; Koyama, N.; Nojima, Y.; Town, T. Overexpression of human S100B exacerbates brain damage and periinfarct gliosis after permanent focal ischemia. Stroke 2008, 39, 2114–2121. [Google Scholar] [CrossRef]
  80. Zhou, S.; Zhu, W.; Zhang, Y.; Pan, S.; Bao, J. S100B promotes microglia M1 polarization and migration to aggravate cerebral ischemia. Inflamm. Res. 2018, 67, 937–949. [Google Scholar] [CrossRef]
  81. Tateishi, N.; Mori, T.; Kagamiishi, Y.; Satoh, S.; Katsube, N.; Morikawa, E.; Morimoto, T.; Matsui, T.; Asano, T. Astrocytic activation and delayed infarct expansion after permanent focal ischemia in rats. Part II: Suppression of astrocytic activation by a novel agent (R)-(-)-2-propyloctanoic acid (ONO-2506) leads to mitigation of delayed infarct expansion and early improvement of neurologic deficits. J. Cereb. Blood Flow Metab. 2002, 22, 723–734. [Google Scholar] [PubMed]
  82. Wajima, D.; Nakagawa, I.; Nakase, H.; Yonezawa, T. Neuroprotective effect of suppression of astrocytic activation by arundic acid on brain injuries in rats with acute subdural hematomas. Brain Res. 2013, 1519, 127–135. [Google Scholar] [CrossRef] [PubMed]
  83. Cordeiro, J.L.; Neves, J.D.; Vizuete, A.F.; Aristimunha, D.; Pedroso, T.A.; Sanches, E.F.; Gonçalves, C.A.; Netto, C.A. Arundic Acid (ONO-2506), an Inhibitor of S100B Protein Synthesis, Prevents Neurological Deficits and Brain Tissue Damage Following Intracerebral Hemorrhage in Male Wistar Rats. Neuroscience 2020, 440, 97–112. [Google Scholar] [CrossRef] [PubMed]
  84. Cordeiro, J.L.; Neves, J.D.; Nicola, F.; Vizuete, A.F.; Sanches, E.F.; Gonçalves, C.A.; Netto, C.A. Arundic Acid (ONO-2506) Attenuates Neuroinflammation and Prevents Motor Impairment in Rats with Intracerebral Hemorrhage. Cell. Mol. Neurobiol. 2022, 42, 739–751. [Google Scholar] [CrossRef] [PubMed]
  85. Kabadi, S.V.; Stoica, B.A.; Zimmer, D.B.; Afanador, L.; Duffy, K.B.; Loane, D.J.; Faden, A.I. S100B inhibition reduces behavioral and pathologic changes in experimental traumatic brain injury. J. Cereb. Blood Flow Metab. 2015, 35, 2010–2020. [Google Scholar] [CrossRef]
  86. Mari, C.; Odorcyk, F.K.; Sanches, E.F.; Wartchow, K.M.; Martini, A.P.; Nicola, F.; Zanotto, C.; Wyse, A.T.; Gonçalves, C.A.; Netto, C.A. Arundic acid administration protects astrocytes, recovers histological damage and memory deficits induced by neonatal hypoxia ischemia in rats. Int. J. Dev. Neurosci. 2019, 76, 41–51. [Google Scholar] [CrossRef]
  87. Higashino, H.; Niwa, A.; Satou, T.; Ohta, Y.; Hashimoto, S.; Tabuchi, M.; Ooshima, K. Immunohistochemical analysis of brain lesions using S100B and glial fibrillary acidic protein antibodies in arundic acid- (ONO-2506) treated stroke-prone spontaneously hypertensive rats. J. Neural Transm. 2009, 116, 1209–1219. [Google Scholar] [CrossRef]
  88. Hanada, M.; Shinjo, R.; Miyagi, M.; Yasuda, T.; Tsutsumi, K.; Sugiura, Y.; Imagama, S.; Ishiguro, N.; Matsuyama, Y. Arundic acid (ONO-2506) inhibits secondary injury and improves motor function in rats with spinal cord injury. J. Neurol. Sci. 2014, 337, 186–192. [Google Scholar] [CrossRef]
  89. Ishiguro, H.; Kaito, T.; Hashimoto, K.; Kushioka, J.; Okada, R.; Tsukazaki, H.; Kodama, J.; Bal, Z.; Ukon, Y.; Takenaka, S.; et al. Administration of ONO-2506 suppresses neuropathic pain after spinal cord injury by inhibition of astrocytic activation. Spine J. 2019, 19, 1434–1442. [Google Scholar] [CrossRef]
  90. Zhang, J.H.; Li, J.K.; Ma, L.L.; Lou, J.Y. RNA interference-mediated silencing of S100B improves nerve function recovery and inhibits hippocampal cell apoptosis in rat models of ischemic stroke. J. Cell. Biochem. 2018, 119, 8095–8111. [Google Scholar] [CrossRef]
  91. Kleindienst, A.; McGinn, M.J.; Harvey, H.B.; Colello, R.J.; Hamm, R.J.; Bullock, M.R. Enhanced hippocampal neurogenesis by intraventricular S100B infusion is associated with improved cognitive recovery after traumatic brain injury. J. Neurotrauma 2005, 22, 645–655. [Google Scholar] [CrossRef]
  92. Baecker, J.; Wartchow, K.; Sehm, T.; Ghoochani, A.; Buchfelder, M.; Kleindienst, A. Treatment with the Neurotrophic Protein S100B Increases Synaptogenesis after Traumatic Brain Injury. Neurotrauma 2020, 37, 1097–1107. [Google Scholar] [CrossRef] [PubMed]
  93. Pitkänen, A.; Löscher, W.; Vezzani, A.; Becker, A.J.; Simonato, M.; Lukasiuk, K.; Gröhn, O.; Bankstahl, J.P.; Friedman, A.; Aronica, E.; et al. Advances in the development of biomarkers for epilepsy. Lancet Neurol. 2016, 15, 843–856. [Google Scholar] [CrossRef] [PubMed]
  94. Liang, K.G.; Mu, R.Z.; Liu, Y.; Jiang, D.; Jia, T.T.; Huang, Y.J. Increased Serum S100B Levels in Patients with Epilepsy: A Systematic Review and Meta-Analysis Study. Front. Neurosci. 2019, 13, 456. [Google Scholar] [CrossRef]
  95. Kumar, P. miRNA dysregulation in traumatic brain injury and epilepsy: A systematic review to identify putative biomarkers for post-traumatic epilepsy. Metab. Brain Dis. 2023, 38, 749–765. [Google Scholar] [CrossRef]
  96. Banote, R.K.; Akel, S.; Zelano, J. Blood biomarkers in epilepsy. Acta Neurol. Scand. 2022, 146, 362–368. [Google Scholar] [CrossRef]
  97. Rubio, T.; Viana, R.; Moreno-Estellés, M.; Campos-Rodríguez, Á.; Sanz, P. TNF and IL6/Jak2 signaling pathways are the main contributors of the glia-derived neuroinflammation present in Lafora disease, a fatal form of progressive myoclonus epilepsy. Neurobiol. Dis. 2023, 176, 105964. [Google Scholar] [CrossRef]
  98. Wang, M.; Yu, J.; Xiao, X.; Zhang, B.; Tang, J. Changes of biochemical biomarkers in the serum of children with convulsion status epilepticus: A prospective study. BMC Neurol. 2022, 22, 196. [Google Scholar] [CrossRef] [PubMed]
  99. Hanin, A.; Denis, J.A.; Frazzini, V.; Cousyn, L.; Imbert-Bismut, F.; Rucheton, B.; Bonnefont-Rousselot, D.; Marois, C.; Lambrecq, V.; Demeret, S.; et al. Neuron Specific Enolase, S100-beta protein and progranulin as diagnostic biomarkers of status epilepticus. J. Neurol. 2022, 269, 3752–3760. [Google Scholar] [CrossRef]
  100. Eriksson, H.; Banote, R.K.; Larsson, D.; Blennow, K.; Zetterberg, H.; Zelano, J. Brain injury markers in new-onset seizures in adults: A pilot study. Seizure 2021, 92, 62–67. [Google Scholar] [CrossRef]
  101. Liang, M.; Zhang, L.; Geng, Z. Advances in the Development of Biomarkers for Post stroke Epilepsy. Biomed. Res. Int. 2021, 2021, 5567046. [Google Scholar] [CrossRef] [PubMed]
  102. Simani, L.; Sadeghi, M.; Ryan, F.; Dehghani, M.; Niknazar, S. Elevated Blood-Based Brain Biomarker Levels in Patients with Epileptic Seizures: A Systematic Review and Meta-analysis.ACS. Chem. Neurosci. 2020, 11, 4048–4059. [Google Scholar] [CrossRef] [PubMed]
  103. Abraira, L.; Santamarina, E.; Cazorla, S.; Bustamante, A.; Quintana, M.; Toledo, M.; Fonseca, E.; Grau-López, L.; Jiménez, M.; Ciurans, J.; et al. Blood biomarkers predictive of epilepsy after an acute stroke event. Epilepsia 2020, 61, 2244–2253. [Google Scholar] [CrossRef] [PubMed]
  104. Kaciński, M.; Budziszewska, B.; Lasoń, W.; Zając, A.; Skowronek-Bała, B.; Leśkiewicz, M.; Kubik, A.; Basta-Kaim, A. Level of S100B protein, neuron specific enolase, orexin A, adiponectin and insulin-like growth factor in serum of pediatric patients suffering from sleep disorders with or without epilepsy. Pharmacol. Rep. 2012, 64, 1427–1433. [Google Scholar] [CrossRef]
  105. Calik, M.; Abuhandan, M.; Sonmezler, A.; Kandemır, H.; Oz, I.; Taskin, A.; Selek, S.; Iscan, A. Elevated serum S-100B levels in children with temporal lobe epilepsy. Seizure 2013, 22, 99–102. [Google Scholar] [CrossRef]
  106. Calik, M.; Abuhandan, M.; Kandemir, H.; Güzel, B.; Solmaz, A.; Celik, H.; Taskin, A.; Iscan, A. Interictal serum S-100B protein levels in intractable epilepsy: A case-control study. Neurosci. Lett. 2014, 558, 58–61. [Google Scholar] [CrossRef]
  107. Meguid, N.A.; Samir, H.; Bjørklund, G.; Anwar, M.; Hashish, A.; Koura, F.; Chirumbolo, S.; Hashem, S.; El-Bana, M.A.; Hashem, H.S. Altered S100 Calcium-Binding Protein B and Matrix Metallopeptidase 9 as Biomarkers of Mesial Temporal Lobe Epilepsy with Hippocampus Sclerosis. J. Mol. Neurosci. 2018, 66, 482–491. [Google Scholar] [CrossRef]
  108. Lu, C.; Li, J.; Sun, W.; Feng, L.; Li, L.; Liu, A.; Li, J.; Mao, W.; Wei, H.; Gao, L.; et al. Elevated plasma S100B concentration is associated with mesial temporal lobe epilepsy in Han Chinese: A case-control study. Neurosci. Lett. 2010, 484, 139–142. [Google Scholar] [CrossRef]
  109. Maiti, R.; Mishra, B.R.; Jena, M.; Mishra, A.; Nath, S.; Srinivasan, A. Effect of anti-seizure drugs on serum S100B in patients with focal seizure: A randomized controlled trial. J. Neurol. 2018, 265, 2594–2601. [Google Scholar] [CrossRef]
  110. Chen, W.; Tan, Y.; Ge, Y.; Chen, Y.; Liu, X. The Effects of Levetiracetam on Cerebrospinal Fluid and Plasma NPY and GAL, and on the Components of Stress Response System, hs-CRP, and S100B Protein in Serum of Patients with Refractory Epilepsy. Cell Biochem. Biophys. 2015, 73, 489–494. [Google Scholar] [CrossRef]
  111. Dahal, A.; Govindarajan, K.; Kar, S. Administration of Kainic Acid Differentially Alters Astrocyte Markers and Transiently Enhanced Phospho-tau Level in Adult Rat Hippocampus. Neuroscience 2023, 516, 27–41. [Google Scholar] [CrossRef] [PubMed]
  112. Somera-Molina, K.C.; Robin, B.; Somera, C.A.; Anderson, C.; Stine, C.; Koh, S.; Behanna, H.A.; Van Eldik, L.J.; Watterson, D.M.; Wainwright, M.S. Glial activation links early-life seizures and long-term neurologic dysfunction: Evidence using a small molecule inhibitor of proinflammatory cytokine upregulation. Epilepsia 2007, 48, 1785–1800. [Google Scholar] [CrossRef]
  113. Vazifehkhah, S.; Khanizadeh, A.M.; Mojarad, T.B.; Nikbakht, F. The possible role of progranulin on anti-inflammatory effects of metformin in temporal lobe epilepsy. J. Chem. Neuroanat. 2020, 109, 101849. [Google Scholar] [CrossRef] [PubMed]
  114. Vizuete, A.F.K.; Hansen, F.; Negri, E.; Leite, M.C.; de Oliveira, D.L.; Gonçalves, C.A. Effects of dexamethasone on the Li-pilocarpine model of epilepsy: Protection against hippocampal inflammation and astrogliosis. J. Neuroinflamm. 2018, 15, 68. [Google Scholar] [CrossRef] [PubMed]
  115. Meng, X.J.; Wang, F.; Li, C.K. Resveratrol is Neuroprotective and Improves Cognition in Pentylenetetrazole-kindling Model of Epilepsy in Rats. Indian J. Pharm. Sci. 2014, 76, 125–131. [Google Scholar]
  116. Vizuete, A.F.K.; Leal, M.B.; Moreira, A.P.; Seady, M.; Taday, J.; Gonçalves, C.A. Arundic acid (ONO-2506) downregulates neuroinflammation and astrocyte dysfunction after status epilepticus in young rats induced by Li-pilocarpine. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2023, 123, 110704. [Google Scholar] [CrossRef]
  117. Dyck, R.H.; Bogoch, I.I.; Marks, A.; Melvin, N.R.; Teskey, G.C. Enhanced epileptogenesis in S100B knockout mice. Brain Res. Mol. Brain Res. 2002, 106, 22–29. [Google Scholar] [CrossRef]
  118. Margolis, K.G.; Gershon, M.D. Enteric neuronal regulation of intestinal inflammation. Trends Neurosci. 2016, 39, 614–624. [Google Scholar] [CrossRef]
  119. Capoccia, E.; Cirillo, C.; Gigli, S.; Pesce, M.; D’Alessandro, A.; Cuomo, R.; Sarnelli, G.; Steardo, L.; Esposito, G. Enteric glia: A new player in inflammatory bowel diseases. Int. J. Immunopathol. Pharmacol. 2015, 28, 443–451. [Google Scholar] [CrossRef]
  120. Seguella, L.; Gulbransen, B.D. Enteric glial biology, intercellular signalling and roles in gastrointestinal disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 571–587. [Google Scholar] [CrossRef]
  121. Ochoa-Cortes, F.; Turco, F.; Linan-Rico, A.; Soghomonyan, S.; Whitaker, E.; Wehner, S.; Cuomo, R.; Christofi, F.L. Enteric Glial Cells: A New Frontier in Neurogastroenterology and Clinical Target for Inflammatory Bowel Diseases. Inflamm. Bowel Dis. 2016, 22, 433–449. [Google Scholar] [CrossRef]
  122. Liñán-Rico, A.; Turco, F.; Ochoa-Cortes, F.; Harzman, A.; Needleman, B.J.; Arsenescu, R.; Abdel-Rasoul, M.; Fadda, P.; Grants, I.; Whitaker, E.; et al. Molecular Signaling and Dysfunction of the Human Reactive Enteric Glial Cell Phenotype: Implications for GI Infection, IBD, POI, Neurological, Motility, and GI Disorders. Inflamm. Bowel Dis. 2016, 22, 1812–1834. [Google Scholar] [CrossRef] [PubMed]
  123. Ferri, G.L.; Probert, L.; Cocchia, D.; Michetti, F.; Marangos, P.J.; Polak, J.M. Evidence for the presence of S-100 protein in the glial component of the human enteric nervous system. Nature 1982, 297, 409–410. [Google Scholar] [CrossRef] [PubMed]
  124. Hao, M.M.; Capoccia, E.; Cirillo, C.; Boesmans, W.; Vanden Berghe, P. Arundic Acid Prevents Developmental Upregulation of S100B Expression and Inhibits Enteric Glial Development. Front. Cell. Neurosci. 2017, 11, 42. [Google Scholar] [CrossRef] [PubMed]
  125. Cirillo, C.; Sarnelli, G.; Esposito, G.; Turco, F.; Steardo, L.; Cuomo, R. S100B protein in the gut: The evidence for enteroglial-sustained intestinal inflammation. World J. Gastroenterol. 2011, 17, 1261–1266. [Google Scholar] [CrossRef]
  126. Celikbilek, A.; Celikbilek, M.; Sabah, S.; Tanık, N.; Borekci, E.; Dogan, S.; Akin, Y.; Baldane, S.; Deniz, K.; Yilmaz, N.; et al. The Serum S100B Level as a Biomarker of Enteroglial Activation in Patients with Ulcerative Colitis. Int. J. Inflam. 2014, 2014, 986525. [Google Scholar] [CrossRef]
  127. Di Liddo, R.; Piccione, M.; Schrenk, S.; Dal Magro, C.; Cosma, C.; Padoan, A.; Contran, N.; Scapellato, M.L.; Pagetta, A.; Romano Spica, V.; et al. S100B as a new fecal biomarker of inflammatory bowel diseases. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 323–332. [Google Scholar] [PubMed]
  128. Cirillo, C.; Sarnelli, G.; Esposito, G.; Grosso, M.; Petruzzelli, R.; Izzo, P.; Calì, G.; D’Armiento, F.P.; Rocco, A.; Nardone, G.; et al. Increased mucosal nitric oxide production in ulcerative colitis is mediated in part by the enteroglial-derived S100B protein. Neurogastroenterol. Motil. 2009, 21, 1209-e112. [Google Scholar] [CrossRef]
  129. Esposito, G.; Capoccia, E.; Sarnelli, G.; Scuderi, C.; Cirillo, C.; Cuomo, R.; Steardo, L. The antiprotozoal drug pentamidine ameliorates experimentally induced acute colitis in mice. J. Neuroinflamm. 2012, 9, 277. [Google Scholar] [CrossRef]
  130. Costa, D.V.S.; Bon-Frauches, A.C.; Silva, A.M.H.P.; Lima-Júnior, R.C.P.; Martins, C.S.; Leitão, R.F.C.; Freitas, G.B.; Castelucci, P.; Bolick, D.T.; Guerrant, R.L.; et al. 5-Fluorouracil Induces Enteric Neuron Death and Glial Activation During Intestinal Mucositis via a S100B-RAGE-NFκB-Dependent Pathway. Sci. Rep. 2019, 9, 665. [Google Scholar] [CrossRef]
  131. Esposito, G.; Capoccia, E.; Turco, F.; Palumbo, I.; Lu, J.; Steardo, A.; Cuomo, R.; Sarnelli, G.; Steardo, L. Palmitoylethanolamide improves colon inflammation through an enteric glia/toll like receptor 4-dependent PPAR-α activation. Gut 2014, 63, 1300–1312. [Google Scholar] [CrossRef] [PubMed]
  132. Turco, F.; Sarnelli, G.; Cirillo, C.; Palumbo, I.; De Giorgi, F.; D’Alessandro, A.; Cammarota, M.; Giuliano, M.; Cuomo, R. Enteroglial-derived S100B protein integrates bacteria-induced Toll-like receptor signalling in human enteric glial cells. Gut 2014, 63, 105–115. [Google Scholar] [CrossRef] [PubMed]
  133. Seguella, L.; Rinaldi, F.; Marianecci, C.; Capuano, R.; Pesce, M.; Annunziata, G.; Casano, F.; Bassotti, G.; Sidoni, A.; Milone, M.; et al. Pentamidine niosomes thwart S100B effects in human colon carcinoma biopsies favouring wtp53 rescue. J. Cell. Mol. Med. 2020, 24, 3053–3063. [Google Scholar] [CrossRef]
  134. Haneishi, Y.; Furuya, Y.; Hasegawa, M.; Picarelli, A.; Rossi, M.; Miyamoto, J. Inflammatory Bowel Diseases and Gut Microbiota. Int. J. Mol. Sci. 2023, 24, 3817. [Google Scholar] [CrossRef] [PubMed]
  135. Orsini, M.; Di Liddo, R.; Valeriani, F.; Mancin, M.; D’Incà, R.; Castagnetti, A.; Aceti, A.; Parnigotto, P.P.; Romano Spica, V.; Michetti, F. In Silico Evaluation of Putative S100B Interacting Proteins in Healthy and IBD Gut Microbiota. Cells 2020, 9, 1697. [Google Scholar] [CrossRef]
  136. Romano Spica, V.; Valeriani, F.; Orsini, M.; Clementi, M.E.; Seguella, L.; Gianfranceschi, G.; Di Liddo, R.; Di Sante, G.; Ubaldi, F.; Ria, F.; et al. S100B Affects Gut Microbiota Biodiversity. Int. J. Mol. Sci. 2023, 24, 2248. [Google Scholar] [CrossRef]
  137. Gazzolo, D.; Monego, G.; Corvino, V.; Bruschettini, M.; Bruschettini, P.; Zelano, G.; Michetti, F. Human milk contains S100B protein. Biochim. Biophys. Acta 2003, 1619, 209–212. [Google Scholar] [CrossRef]
  138. Galvano, F.; Frigiola, A.; Gagliardi, L.; Ciotti, S.; Bognanno, M.; Iacopino, A.M.; Nigro, F.; Tina, G.L.; Cavallaro, D.; Mussap, M.; et al. S100B milk concentration in mammalian species. Front. Biosci. 2009, 1, 542–546. [Google Scholar]
  139. Skaper, S.D.; Facci, L.; Zusso, M.; Giusti, P. An Inflammation-Centric View of Neurological Disease: Beyond the Neuron. Front. Cell. Neurosci. 2018, 12, 72. [Google Scholar] [CrossRef] [PubMed]
  140. Mao, D.; Zheng, Y.; Xu, F.; Han, X.; Zhao, H. HMGB1 in nervous system diseases: A common biomarker and potential therapeutic target. Front. Neurol. 2022, 13, 1029891. [Google Scholar] [CrossRef]
  141. Singh, H.; Agrawal, D.K. Therapeutic Potential of Targeting the HMGB1/RAGE Axis in Inflammatory Diseases. Molecules 2022, 27, 7311. [Google Scholar] [CrossRef] [PubMed]
  142. Shi, Y.; Zhu, N.; Qiu, Y.; Tan, J.; Wang, F.; Qin, L.; Dai, A. Resistin-like molecules: A marker, mediator and therapeutic target for multiple diseases. Cell Commun. Signal. 2023, 21, 18. [Google Scholar] [CrossRef] [PubMed]
  143. Hudson, B.I.; Lippman, M.E. Targeting RAGE Signaling in Inflammatory Disease. Annu. Rev. Med. 2018, 69, 349–364. [Google Scholar] [CrossRef] [PubMed]
  144. Hartman, K.G.; McKnight, L.E.; Liriano, M.A.; Weber, D.J. The evolution of S100B inhibitors for the treatment of malignant melanoma. Future Med. Chem. 2013, 5, 97–109. [Google Scholar] [CrossRef]
  145. Cristóvão, J.S.; Romão, M.A.; Gallardo, R.; Schymkowitz, J.; Rousseau, F.; Gomes, C.M. Targeting S100B with Peptides Encoding Intrinsic Aggregation-Prone Sequence Segments. Molecules 2021, 26, 440. [Google Scholar] [CrossRef] [PubMed]
  146. Ivanenkov, V.V.; Jamieson GAJr Gruenstein, E.; Dimlich, R.V. Characterization of S-100b binding epitopes. Identification of a novel target, the actin capping protein, CapZ. J. Biol. Chem. 1995, 270, 14651–14658. [Google Scholar] [CrossRef]
  147. Kannan, S.; Aronica, P.G.A.; Nguyen, T.B.; Li, J.; Verma, C.S. Computational Design of Macrocyclic Binders of S100B(ββ): Novel Peptide Theranostics. Molecules 2021, 26, 721. [Google Scholar] [CrossRef]
  148. Young, B.D.; Yu, W.; Rodríguez, D.J.V.; Varney, K.M.; MacKerell, A.D., Jr.; Weber, D.J. Specificity of Molecular Fragments Binding to S100B versus S100A1 as Identified by NMR and Site Identification by Ligand Competitive Saturation (SILCS). Molecules 2021, 26, 381. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of effects induced by extracellular astrocytic S100B. At nanomolar concentrations, S100B does not impact the homeostasis of microglia. In contrast, when S100B reaches micromolar concentrations, microglia rapidly activate NF-κB-dependent transcription and exhibit pro-inflammatory phenotypes. Negative regulation of extracellular S100B can reprogram microglia from inflammation towards homeostasis via suppression of NF-κB signaling.
Figure 1. Schematic representation of effects induced by extracellular astrocytic S100B. At nanomolar concentrations, S100B does not impact the homeostasis of microglia. In contrast, when S100B reaches micromolar concentrations, microglia rapidly activate NF-κB-dependent transcription and exhibit pro-inflammatory phenotypes. Negative regulation of extracellular S100B can reprogram microglia from inflammation towards homeostasis via suppression of NF-κB signaling.
Ijms 24 09605 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Michetti, F.; Clementi, M.E.; Di Liddo, R.; Valeriani, F.; Ria, F.; Rende, M.; Di Sante, G.; Romano Spica, V. The S100B Protein: A Multifaceted Pathogenic Factor More Than a Biomarker. Int. J. Mol. Sci. 2023, 24, 9605. https://doi.org/10.3390/ijms24119605

AMA Style

Michetti F, Clementi ME, Di Liddo R, Valeriani F, Ria F, Rende M, Di Sante G, Romano Spica V. The S100B Protein: A Multifaceted Pathogenic Factor More Than a Biomarker. International Journal of Molecular Sciences. 2023; 24(11):9605. https://doi.org/10.3390/ijms24119605

Chicago/Turabian Style

Michetti, Fabrizio, Maria Elisabetta Clementi, Rosa Di Liddo, Federica Valeriani, Francesco Ria, Mario Rende, Gabriele Di Sante, and Vincenzo Romano Spica. 2023. "The S100B Protein: A Multifaceted Pathogenic Factor More Than a Biomarker" International Journal of Molecular Sciences 24, no. 11: 9605. https://doi.org/10.3390/ijms24119605

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

Article Metrics

Back to TopTop