Next Article in Journal
Development of a Method for the In Vivo Generation of Allogeneic Hearts in Chimeric Mouse Embryos
Next Article in Special Issue
Role of Inflammatory/Immune Response and Cytokine Polymorphisms in the Severity of Chronic Hepatitis C (CHC) before and after Direct Acting Antiviral (DAAs) Treatment
Previous Article in Journal
Physiological, Biochemical, and Epigenetic Reaction of Maize (Zea mays L.) to Cultivation in Conditions of Varying Soil Salinity and Foliar Application of Silicon
Previous Article in Special Issue
LPS-Induced Inhibition of miR-143 Expression in Brown Adipocytes Promotes Thermogenesis and Fever
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumor Necrosis Factor (TNF)-α-Stimulated Gene 6 (TSG-6): A Promising Immunomodulatory Target in Acute Neurodegenerative Diseases

1
Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
2
Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, and Instituto de Investigación Hospital 12 de Octubre (Imas12), 28040 Madrid, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(2), 1162; https://doi.org/10.3390/ijms24021162
Submission received: 5 December 2022 / Revised: 26 December 2022 / Accepted: 4 January 2023 / Published: 6 January 2023
(This article belongs to the Special Issue Regulation of Inflammatory Reactions in Health and Disease 2.0)

Abstract

:
Tumor necrosis factor (TNF)-α-stimulated gene 6 (TSG-6), the first soluble chemokine-binding protein to be identified in mammals, inhibits chemotaxis and transendothelial migration of neutrophils and attenuates the inflammatory response of dendritic cells, macrophages, monocytes, and T cells. This immunoregulatory protein is a pivotal mediator of the therapeutic efficacy of mesenchymal stem/stromal cells (MSC) in diverse pathological conditions, including neuroinflammation. However, TSG-6 is also constitutively expressed in some tissues, such as the brain and spinal cord, and is generally upregulated in response to inflammation in monocytes/macrophages, dendritic cells, astrocytes, vascular smooth muscle cells and fibroblasts. Due to its ability to modulate sterile inflammation, TSG-6 exerts protective effects in diverse degenerative and inflammatory diseases, including brain disorders. Emerging evidence provides insights into the potential use of TSG-6 as a peripheral diagnostic and/or prognostic biomarker, especially in the context of ischemic stroke, whereby the pathobiological relevance of this protein has also been demonstrated in patients. Thus, in this review, we will discuss the most recent data on the involvement of TSG-6 in neurodegenerative diseases, particularly focusing on relevant anti-inflammatory and immunomodulatory functions. Furthermore, we will examine evidence suggesting novel therapeutic opportunities that can be afforded by modulating TSG-6-related pathways in neuropathological contexts and, most notably, in stroke.

1. Introduction

Stroke is a leading cause of mortality and disability worldwide, and implementation of prevention and treatment strategies is imperative to limit the expected growth of its global burden [1]. To date, the only available therapeutic strategy for acute ischemic stroke consists in thrombus lysis/removal achieved by intravenous administration of human recombinant tissue plasminogen activator (rt-PA) or other proteins with similar activity [2,3,4] and/or endovascular thrombectomy or embolectomy [5,6,7,8]. These interventions are based on reperfusion of the infarcted area; while they do not exert direct neuroprotection to block the progression of brain damage, that remains a hopeful target. To this end, in the last several decades, substantial progress has been made to delineate the complex molecular processes underlying the pathobiological mechanisms implicated in ischemic brain damage [9,10]. Decreased blood flow to the brain triggers an ischemic detrimental cascade involving excitotoxicity, oxidative stress, and inflammation that contribute to the development of cerebral damage [11,12,13]. Moreover, several studies support the evidence that central and peripheral immune responses play a pivotal role in ischemic pathobiology, exerting detrimental or protective functions, depending on the specific molecules or cells (phenotypes/subtypes) involved, as well as on the phases after the initial insult [14,15,16]. To implement therapeutic approaches, recent experimental work has suggested that an appropriate immunomodulation would allow to achieve neuroprotection. Thus, dissecting the molecular mechanisms by which the immune system is implicated in ischemic stroke injury is crucial for the identification of novel therapeutic targets. To this end, in this review, we will discuss the most recent data on the involvement of tumor necrosis factor (TNF)-α-stimulated gene 6 (TSG-6) in acute and chronic neurodegenerative diseases, particularly focusing on relevant anti-inflammatory and immunomodulatory functions. Furthermore, we will examine evidence suggesting novel therapeutic opportunities that can be afforded by modulating TSG-6-related pathways in neuropathological contexts and, most notably, in stroke.

2. Inflammatory and Immune Mechanisms in Ischemic Stroke

Damage-associated molecular pattern molecules (DAMPs), released following ischemic cell death, trigger inflammatory responses by interacting with pattern recognition receptors, including nucleotide oligomerization domain (NOD)-like receptors, receptor of advanced glycation end-products (RAGE), and Toll-like receptors (TLRs), the latter being involved in the initiation of innate immunity [17,18,19]. Upon ligand binding, TLR4 dimerizes and promotes myeloid differentiation primary response (MyD)88- or TIR-domain-containing adapter-inducing interferon-β (TRIF)-dependent signaling, implicated in downstream activation of nuclear factor kappa B (NF-κB) that is, in turn, necessary for inflammatory cytokine production [20,21].
After sensing tissue damage, resting ramified microglia gradually transform into amoeboid-shaped cells that initially clear debris and foster tissue repair [22,23,24]. These ‘beneficial’ functions, typically provided by M2-like phenotypes, are rapidly replaced by a transition toward pathological M1 subsets [25] that express a range of inflammatory mediators, such as TNF-α, interleukin (IL)-1β, IL-6, inducible nitric oxide synthase (iNOS), monocyte chemoattractant protein (MCP)-1, and macrophage inflammatory protein (MIP)-1α [26,27,28,29,30]. Indeed, a range of diverse transcriptional programs have been reported to occur in rodent and human microglia, since, in the late stages, M2-like phenotypes release proresolving cytokines (i.e., IL-10 and transforming growth factor-β, TGF-β) and trophic factors that exert proangiogenic and reparative functions [31,32,33,34,35].
Stroke-induced sterile inflammation also involves mobilization and cerebral recruitment of circulating monocytes, neutrophils, and T cells [36,37,38,39,40]. Regarding monocytes, classical CD14+/CD16- subtypes mainly secrete inflammatory TNF-α, IL-6 and IL-1β [41,42,43], and their blood number, as well as their expression of TLR-4, increase early after stroke, being independently associated with poor outcome in patients [44,45,46]. Moreover, higher monocyte counts positively correlate with ischemic stroke severity and adverse prognosis [47,48,49], whereas the less represented CD16+ intermediate and nonclassical monocytes have been found to be inversely related to poor outcomes and mortality, respectively [44], likely exerting protective/reparative functions [50]. Once recruited in the injured tissue, bloodborne monocytes transform into subsets of macrophages, as shown both in patients and in rodent models [51,52,53].
In the early stage after ischemia, alternatively-activated M2 subsets predominate in the ischemic core region, where they display reparative functions [54]. Thereafter, they switch toward M1-like phenotypes that abound in the injured brain to elicit detrimental effects through the release of inflammatory and toxic mediators, including TNF-α, IL-1β, and ROS [25,29,35,40,55,56,57].
The dualistic role of innate immunity in ischemic pathophysiology is further underscored by ability of neutrophils to adopt N1 inflammatory or N2 beneficial phenotypes by TLR4-dependent mechanisms among other mechanisms [58,59,60,61]. Early after injury, they release inflammatory and neurotoxic mediators, including cytokines, ROS and proteases that prompt BBB damage, tissue edema, hemorrhagic transformation, and cerebral damage [59,62]. Once recruited to the ischemic tissue, neutrophils contribute to thrombus formation and expansion and impair revascularization and vascular remodeling through the release of neutrophil extracellular traps (NETs) [63,64]. Indeed, increased blood or brain-infiltrating neutrophils are associated with stroke severity and poor functional outcome [65]. However, the attempts to block these detrimental immune cells resulted in limited therapeutic success [59,65], likely due to the beneficial roles of N2 phenotypes that facilitate macrophage phagocytosis and may support neuroprotection [66,67,68].
A recent bioinformatic study revealed that among key immunoregulatory pathways implicated in ischemic stroke pathogenesis, TNF-related signaling covers a pivotal role [69]. TNF is an inflammatory cytokine with dualistic effects in the brain [70,71], since its soluble form is detrimental, whereas its membrane-bound form displays protective properties and is required for the maintenance of innate immunity [72,73,74,75,76]. After either experimental or human stroke, cerebral expression of TNF and of its receptors, TNFR1 and TNFR2, increases [77,78,79,80,81], whereby microglia and infiltrating macrophages represent the major source of TNF-α in the acute phase after ischemia [80,82]. Intriguingly, TNF-α mRNA levels are elevated in circulating monocytes isolated from ischemic stroke patients 24–48 h after symptoms onset [83], and both TNFR1 and TNFR2 are differentially regulated in distinct subpopulations of monocytes and neutrophils [84].

3. Biological Effects of TSG-6

Among the multitude of genes regulated by TNF-α is the TNF-α-induced protein 6 (TNFAIP6) gene, coding for the secretory protein TSG-6, initially identified in human fibroblasts lines and peripheral blood mononuclear cells [85,86]. TSG-6 can be induced by diverse cytokines (i.e., TNF, IL-1) and other inflammatory stimuli (i.e., LPS) and is a member of the family of hyaluronate (HA) binding proteins involved in cell–cell and cell–matrix interactions during inflammation and tumorigenesis [85,87,88]. In fact, although constitutively expressed in some tissues, including the brain and spinal cord, TSG-6 is generally upregulated in response to inflammation in a wide variety of cell types, such as monocytes/macrophages, dendritic cells, astrocytes, mesenchymal stem/stromal cells (MSCs), vascular smooth muscle cells (VSMCs), and fibroblasts [89,90,91,92,93]. TSG-6 has a variety of activities, including the modulation of immune and stromal cells function and the regulation of extracellular matrix organization and interaction with cell surface receptors and soluble mediators (e.g., chemokines) (figure in Section 4) [94]. The anti-inflammatory functions of TSG-6 rely on direct modulation of inflammatory cells and on the regulation of the assembly/organization of HA matrices that underlie its immunosuppressive properties [95,96,97,98,99,100,101,102]. TSG-6 was also shown to inhibit NETs release from bone-marrow-derived neutrophils [103]. Moreover, the original evidence that TSG-6 produced by MSCs underlies their immunomodulatory and reparative functions [104,105] has stimulated interest in understanding the role of this molecule in a number of pathological conditions. Indeed, an increasing body of experimental evidence suggests that the main function of this protein is to protect tissue from the damaging effects caused by inflammation, as demonstrated in animal models of acute myocardial infarction [104], atherosclerosis [106], lung injury [107,108], arthritis [109,110,111,112,113], spinal cord injury [114], and, most notably, acute brain injury [115,116,117,118,119,120].

4. Immunomodulatory Functions of TSG-6 In Vitro

TSG-6 was the first soluble chemokine-binding protein to be identified in mammals, as it binds to a wide range of chemokines belonging to both CC and CXC subfamilies, hindering their presentation and interaction with matrix molecules [121,122]. By interacting with CXCL8, TSG-6 blocks its binding to endothelial heparan sulphate (HS), a mechanism implicated in its potent (in vitro and in vivo) inhibitory activity on neutrophil chemotaxis and transendothelial migration (Figure 1) [121,122,123,124,125]. The latter mechanism has been reported to underlie the protective effects of TSG-6 in a number of degenerative and inflammatory experimental conditions, including acute brain injury [104,105,117,126,127,128]. Moreover, binding to other chemokines that can be recognized by diverse leukocytes (e.g., CCR5, CCR7, and CXCR4) strongly suggests that TSG-6 may regulate recruitment and migration of different white blood cells (Figure 1) [121,122]. Accordingly, TSG-6 was reported to regulate the function of dendritic cells, macrophages, monocytes, and T cells attenuating their inflammatory responses and promoting immune tolerance [96,104,107,127,129,130,131,132,133].
In human neutrophils, TSG-6 is constitutively present in the secretory lactoferrin-positive granules; monocytes, macrophages, myeloid dendritic cells, and neutrophils produce high levels of TSG-6 in response to inflammatory triggers (LPS or cytokines) [134]. Conversely, anti-inflammatory cytokines, namely IL-4 and IL-10, dampen LPS-induced TSG-6 expression in human leukocytes [134].
In turn, TSG-6 acts in an autocrine mode on macrophages to promote their transition from proinflammatory toward anti-inflammatory phenotypes (Figure 1) [107,133,135]. In particular, this immunomodulatory protein inhibits the association between TLR-4 and MyD88, thus suppressing the activation of NF-κB, and prevents the expression of pro-inflammatory mediators (e.g., IL-6, TNF-α, IL-1β and inducible nitric oxide synthase, iNOS, STAT1, and STAT3), while elevating the expression of anti-inflammatory proteins (e.g., CD206, IL-4, and IL-10) [96,98,107,129,136,137].
The majority of the studies performed to date on TSG-6 were focused on its ability to mediate the therapeutic efficacy of MSC in a number of pathological conditions, including neuroinflammation, since it plays a major role in the modulation of sterile inflammation [94,138]. Bone MSCs stimulated with TNF-α release TSG-6 that has been reported to inhibit production of inflammatory cytokines triggered by LPS in cultured rat astrocytes [115,139]. This effect was mediated by inhibition of NF-κB signaling pathway and was suggested to underlie the protective role of bone NSCs on BBB damage induced by intracerebral or subarachnoid hemorrhage in rat [115,139].
Similarly, MSCs and adipose-derived stem cells (ADSCs) exposed to TNF-α increased their expression of TSG-6 that, once secreted, inhibited the release of inflammatory mediators (i.e., IL-1β, IL-6, TNF-α, iNOS) by cultured BV2 microglia [140,141]. This effect was shown to be mediated by the interaction of TSG-6 with CD44 and subsequent blockade of NF-κB inflammatory signaling in BV2 microglia [141]. In fact, TSG-6 binds and forms stable complexes with HA, thus stabilizing its interaction with the membrane receptor CD44, and resulting in negative regulation of TLR-4-dependent signaling [95,97,142,143]. Accordingly, TSG-6 downregulated the TLR2/MyD88/NF-κB signaling (Figure 1) and reduced the production of proinflammatory cytokines, such as IL-1β, IL-6, and TNF-α, in primary microglia treated with a specific TLR2 agonist, an effect that was suggested to underlie the ability of TSG-6 to attenuate neuropathic pain caused by chronic constriction injury in rats [144]. A similar mechanism was described in cultured murine macrophages, whereby TSG-6 released by stimulated MSCs downregulated TLR2-mediated nuclear translocation of NF-κB, by binding CD44 directly or through a complex with HA [98]. The ability of TSG-6 to inhibit proliferation and release of inflammatory mediators was also suggested to occur through reduced activation of p38 and JNK signaling in rat macrophages [145,146].
TSG-6 also suppressed LPS-induced TNF-α production and the expression of the inflammatory M1 phenotype in human monocyte-derived macrophages [147], while it prevented NF-κB activation by inhibiting the association of MyD88 with TLR4 in mouse macrophages (Figure 1) [107]. Interestingly, TLR2-related pathways were reported to play a major role in the regulation of the polarization state of microglia, while NF-κB and p38 were implicated in polarization of microglia/macrophages during ischemic stroke injury [148,149,150,151,152,153]. This strongly supports the hypothesis that TSG-6 may affect M1 vs. M2 polarization shift of myeloid cells under neuroinflammatory conditions. In line with this speculation is the evidence that BV2 microglia treated with MSCs displayed reduced LPS-induced expression of early and late markers of M1 phenotype (iNOS, IL-1β, CD16, CD86), and concomitant elevation of typical markers of M2 polarization (CD206, Arg1) [154,155]. Notably, most of these effects were lost when LPS-primed BV2 microglia were exposed to MSCs in which TSG-6 was knocked down [154]. In this context, TSG-6 was suggested to modulate microglia polarization shift by preventing LPS-induced phosphorylation of STAT3 [154]. The inhibitory effect of TSG-6 on STAT3 phosphorylation was also demonstrated to underlie polarization toward M2 phenotype in murine macrophages, thus underlying protection in mice undergone LPS-induced inflammatory lung injury or liver inflammation due to alcoholic hepatitis [107,156].

5. TSG-6 as Mediator of the Neuroprotective Functions of MSCs

The anti-inflammatory and immunoregulatory functions of TSG-6 have been studied in a number of neurological disorders (Figure 2), although most studies mainly focused on the role of this multifunctional protein in the protective effects of MSCs, while little is known on its direct mechanisms and on its functions when released from endogenous sources.
In global cerebral ischemia caused by cardiac arrest in rat, intravenous administration of MSCs significantly reduced brain damage by upregulating cerebral expression of TSG-6, while attenuating the ischemia-induced elevation of neutrophil elastase and of inflammatory cytokines (i.e., IL-1β, IL-6, and TNF-α) expression [119,157]. Although administration of recombinant TSG-6 in the lateral ventricle was effective in reducing histological and functional deficits associated with global ischemia, these authors did not investigate the mechanisms involved in cerebral upregulation of TSG-6 by MSCs, but they hypothesized that TSG-6 was actually released by MSCs migrating to the brain [119]. In fact, intravenous infusion of MSCs, in which TSG-6 expression was silenced by siRNA, failed to attenuate brain inflammation in ischemic rats [157].
Interestingly, bone marrow-derived MSCs are effective in ameliorating stroke outcomes and, despite limited knowledge of their exact neuroprotective mechanisms, their immune-suppressive and anti-inflammatory effects are likely to play a crucial role. This is supported by the evidence that intravenous injection of MSCs in mice reduced the elevation of brain and blood levels of the inflammatory complement component C3 induced by focal cerebral ischemia, thus resulting in reduced infarct volume [158]. These effects were also associated with elevation of brain and blood levels of TSG-6, 8 h after MSCs administration, strongly suggesting its involvement in their protective effects [158].
The ability of TSG-6 to attenuate neuroinflammatory reactions was also demonstrated in rats exposed to collagenase to induce intracerebral hemorrhage (ICH), intravenously transplanted with MSCs. In this model, the protective effects of MSCs on BBB and their anti-inflammatory effects were ascribed to the increased mRNA and protein expression of TSG-6 detected in the brain 24 h and 48 h after ICH [159]. In particular, although not directly demonstrated, TSG-6 elevation was suggested to underlie suppression of the activation of NF-kB signaling pathway and downstream reduction of iNOS and peroxynitrite levels [159]. More recent studies have confirmed the ability of bone MSCs to reduce neurological deficits and BBB damage caused by ICH or SAH in rat, through TSG-6 release, since these protective effects were abolished after silencing TSG-6 by siRNA [115,139]. In particular, Tang et al. (2021) speculated that TSG-6, likely secreted via exosomes by bone MSCs, acts through a paracrine mechanism to regulate activated astrocytes to preserve BBB integrity. In addition, TSG-6 was demonstrated to mediate the anti-inflammatory effects of bone MSCs, by inhibiting NF-κB signaling pathways and the cerebral increase of inflammatory cytokines (e.g., IL-1β, IL-6, INF-γ) and peroxynitrite caused by ICH or SAH in rats [115,139].
Enhanced expression of TSG-6 and downstream suppression of NF-κB signaling pathways have also been suggested to underlie the anti-inflammatory effects of MSCs or NS309 (a small conductance Ca2+-activated K+ channels activator) in rodent models of traumatic brain injury (TBI). In fact, intravenous MSCs transplantation 2 h after TBI, or i.p. administration of NS309 30 min before TBI, caused upregulation of TSG-6 in the injured cortex up to 72 h after the insult in rat [120,160]. This was associated with reduced levels of inflammatory cytokines (i.e., IL-1β, IL-6, IL-17, TNF-α, INF-γ) and chemokines (i.e., MCP-1, MIP-2, RANTES), lower density of inflammatory microglia/macrophages and peripheral infiltrating leukocytes, together with elevated levels of anti-inflammatory cytokines (i.e., IL-10, IL-4, TGF-β1) in the lesioned tissue [120,160]. Notably, knockdown of TSG-6 using in vivo transfection with TSG-6 specific shRNA partially reversed the protective and anti-inflammatory effects of NS309 against TBI [160]. To further support its beneficial effects, there is the evidence that intravenous treatment with TSG-6 decreased neutrophil extravasation, matrix metalloproteinase (MMP)-9 expression and the resulting BBB leakage caused by TBI in mice [117]. Remarkably, acute administration of TSG-6 within 24 h of TBI not only reduced brain lesion size at two weeks, but it also promoted neurogenesis and attenuated long-term consequences of TBI, such as memory impairments and depressive-like behavior [117].

6. Roles of Endogenous TSG-6 in Neurodegeneration

Although most studies have focused on TSG-6 as pivotal mediator of the beneficial effects of MSCs, there is also evidence of the role of this multifunctional glycoprotein as endogenous immunoregulatory and anti-inflammatory mediator in brain pathological conditions. TSG-6 is physiologically expressed in the developing rat brain, displaying different expression patterns in distinct cerebral regions where it may play a role in oligodendrocyte maturation and neuronal precursor cell migration [161]. Although its expression during embryonic development is still controversial, other findings have demonstrated the presence of TSG-6 in astrocytes of the mature rat brain and spinal cord [92]. In fact, TSG-6 has been implicated in astrocyte maturation, since fewer GFAP+ astrocytes were found in the brain of TSG-6 null mice [92]. Despite a selective localization of TSG-6 in astrocytes was suggested by some studies, other findings have shown a more widespread distribution of this multifunctional protein.

6.1. Ischemic Stroke

In the brain of stroke patients, both mRNA and protein levels of TSG-6 were elevated in the peri-infarct and infarcted tissue as compared to contralateral hemisphere, being its positive staining associated with inflammatory mononuclear cells and damaged neurons in patients surviving from 3 to 29 days after stroke [162]. This was coincident with elevation of HA levels in infarcted brain tissue and serum of patients up to 37 days after stroke; concomitantly, increased expression of the HA receptor CD44 was mainly observed in dead or dying neurons from infarct or peri-infarct tissue, as well as in inflammatory mononuclear cells 3–17 days after the initial injury [162]. Increased HA synthesis and up-regulation of CD44 in microglia, macrophages, and microvessels of the ischemic brain tissue were also observed in rodents subjected to MCAo [163,164]. The preferential synthesis of high molecular weight HA in stroke tissue, together with the elevation of TSG-6, are likely aimed at modulating inflammatory responses and at re-establishing the extracellular matrix integrity during tissue remodeling after ischemic stroke [162,163,164]. According to human findings, the expression of TSG-6 was also elevated in the cerebral cortex of rats 3 days after global cerebral ischemia due to transient cardiac arrest [119]. Although the elevation of TSG-6 has been suggested to underlie its putative beneficial functions, evidence on the very early phases after ischemic stroke injury is lacking, being a time-course analysis crucial for clarifying the exact role of this multifunctional protein.
The elevation of TSG-6 is not only restricted to the ischemic brain, since recent evidence has highlighted that noncardioembolic acute ischemic stroke patients display higher plasma TSG-6 levels than control subjects [165]. Plasma TSG-6 level positively correlated with stroke severity at admission, lesion volume, neutrophil count, neutrophil-to-lymphocyte ratio, and interleukin-8 level. In those patients, elevated TSG-6 plasma levels were independently associated with three-month poor prognosis, whereas elevated TSG-6 to IL-8 ratio predicted favorable outcome at three months [165]. This latter finding adds complexity to the interpretation of TSG-6 elevation in the periphery, highlighting that the prognostic value of circulating TSG-6 levels needs to be further evaluated. Elevation of circulating levels of TSG-6 was also observed in patients with carotid stenosis or with coronary artery disease, likely deriving from monocyte-derived macrophages, endothelial, and arterial smooth muscle cells exposed to inflammatory stimuli [147,166]. Indeed, both animal and human studies have reported expression of TSG-6 in atherosclerotic lesions, strongly suggesting an atheroprotective role of this anti-inflammatory protein in vascular lesions [106,147,167,168,169]. Intriguingly, high Alzheimer’s disease neuropathologic change (ADNC) and moderate–severe cerebral amyloid angiopathy (CAA), in the absence of concurrent pathologies (e.g., infarctions, Lewy bodies), were associated with increased TSG-6 expression and HA content in cerebral microvascular lysates [170]. In high ADNC subjects, TSG-6 was not only expressed in the vasculature, but also in neurons and astrocytes that concomitantly express HA synthase 2, strongly suggesting the involvement of HA/TSG-6 interplay in the regulation of brain pathological processes [171].

6.2. Other Brain Injuries

Either brain or spinal cord injury resulted in considerable upregulation of TSG-6 mRNA expression, whereby the protein is associated with the glial scar, likely playing a role in formation and stabilization of this HA-rich matrix forming an immunosuppressive environment [92,172]. Upon binding to inflammatory cells, these TSG-6 modified HA matrices modulate their responses, thus contributing to pathological inflammation [100,101]. Accordingly, elevation of endogenous TSG-6 mRNA was recently found in the lesioned hemisphere of mice subjected to penetrating brain injury (PBI), an effect that was associated with an anti-inflammatory role of this protein in the glial scar [172]. Indeed, TSG-6 null mice display a more severe inflammatory response (i.e., higher levels of NF-κB, RANTES and IL-1β, as well as higher number of CD68+ activated microglia and macrophages) and increased glial scar deposition in the injured brain as compared to littermate control mice [172].
In a rat model of SAH, both mRNA and protein levels of TSG-6 were upregulated from 12 to 72 h after injury in the brain, with a prevalent localization in Iba1 immunopositive microglia, where TSG-6 triggers an anti-inflammatory protective phenotype through pSTAT3 regulation [116]. In fact, knockdown of endogenous TSG-6 by siRNA elevated (CD86+) M1 vs. (CD163+) M2 ratio in cerebral microglia and aggravated neurological deficits 24 h after SAH. Conversely, intracerebroventricular (i.c.v.) administration of recombinant TSG-6, 1.5 h after SAH in rat, reduced microglia shift toward inflammatory phenotypes, attenuated TNF-α expression level, and upregulated IL-10 expression levels, thus resulting in reduced brain edema and improved neurological deficit [116,173].
The relevance of inflammatory triggers to induce TSG-6 expression is also highlighted by the evidence that elevation of TSG-6 gene expression was observed in the neonatal brain 4 h after intraperitoneal injection of LPS, where it was suggested to represent a compensatory anti-inflammatory mechanism [161]. In fact, treatment with recombinant TSG-6 exerted neuroprotection by reducing systemic inflammatory responses and cerebral apoptosis caused by LPS in newborn rats [161].

7. Conclusions

Elevation of endogenous TSG-6 has been reported to occur upon diverse acute and chronic neurodegenerative conditions, as a compensatory response aimed at triggering anti-inflammatory and a multitude of (neuro)protective mechanisms. Thus, emerging evidence provides insights into the potential use of TSG-6 as a peripheral biomarker for diagnostic and/or prognostic purposes, especially in the context of ischemic stroke, where the pathobiological relevance of this protein was also validated in patients. Moreover, TSG-6 mediates most of the beneficial functions of MSCs in acute neurodegenerative conditions, including cerebral ischemia, and is itself able to provide neuroprotection when administered to animal models. All these findings strongly highlight the promising efficacy of TSG-6 replacement therapy, with either the peptide itself or its analogues, particularly against acute neurodegenerative insults, and pose the basis for further investigation aimed at characterizing its pharmacokinetic and pharmacodynamic properties in these pathological contexts.

Author Contributions

Conceptualization, D.L.R. and D.A.; writing—original draft preparation, D.L.R. and C.D.S.; writing—review and editing, I.L., A.M., G.B. and D.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Italian Ministry of Education, University and Research (PRIN codes 2015KRYSJN to DA and 2017XKJTLW_001 to GB).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Feigin, V.L.; Stark, B.A.; Johnson, C.O.; Roth, G.A.; Bisignano, C.; Abady, G.G.; Abbasifard, M.; Abbasi-Kangevari, M.; Abd-Allah, F.; Abedi, V.; et al. Global, regional, and national burden of stroke and its risk factors, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol. 2021, 20, 795–820. [Google Scholar] [CrossRef] [PubMed]
  2. Thomalla, G.; Boutitie, F.; Ma, H.; Koga, M.; Ringleb, P.; Schwamm, L.H.; Wu, O.; Bendszus, M.; Bladin, C.F.; Campbell, B.C.V.; et al. Intravenous alteplase for stroke with unknown time of onset guided by advanced imaging: Systematic review and meta-analysis of individual patient data. Lancet 2020, 396, 1574–1584. [Google Scholar] [CrossRef] [PubMed]
  3. Ma, H.; Campbell, B.C.; Parsons, M.W.; Churilov, L.; Levi, C.R.; Hsu, C.; Kleinig, T.J.; Wijeratne, T.; Curtze, S.; Dewey, H.M.; et al. Thrombolysis Guided by Perfusion Imaging up to 9 Hours after Onset of Stroke. N. Engl. J. Med. 2019, 380, 1795–1803. [Google Scholar] [CrossRef] [PubMed]
  4. Hacke, W.; Kaste, M.; Bluhmki, E.; Brozman, M.; Dávalos, A.; Guidetti, D.; Larrue, V.; Lees, K.R.; Medeghri, Z.; Machnig, T.; et al. Thrombolysis with Alteplase 3 to 4.5 Hours after Acute Ischemic Stroke. N. Engl. J. Med. 2008, 359, 1317–1329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Shafie, M.; Yu, W. Recanalization Therapy for Acute Ischemic Stroke with Large Vessel Occlusion: Where We Are and What Comes Next? Transl. Stroke Res. 2021, 12, 369–381. [Google Scholar] [CrossRef]
  6. Albers, G.W.; Marks, M.P.; Kemp, S.; Christensen, S.; Tsai, J.P.; Ortega-Gutierrez, S.; McTaggart, R.A.; Torbey, M.T.; Kim-Tenser, M.; Leslie-Mazwi, T.; et al. Thrombectomy for Stroke at 6 to 16 Hours with Selection by Perfusion Imaging. N. Engl. J. Med. 2018, 378, 708–718. [Google Scholar] [CrossRef]
  7. Berkhemer, O.A.; Fransen, P.S.S.; Beumer, D.; Berg, L.A.V.D.; Lingsma, H.F.; Yoo, A.J.; Schonewille, W.J.; Vos, J.A.; Nederkoorn, P.J.; Wermer, M.J.H.; et al. A Randomized Trial of Intraarterial Treatment for Acute Ischemic Stroke. N. Engl. J. Med. 2015, 372, 11–20. [Google Scholar] [CrossRef] [Green Version]
  8. Nogueira, R.G.; Jadhav, A.P.; Haussen, D.C.; Bonafe, A.; Budzik, R.F.; Bhuva, P.; Yavagal, D.R.; Ribo, M.; Cognard, C.; Hanel, R.A.; et al. Thrombectomy 6 to 24 Hours after Stroke with a Mismatch between Deficit and Infarct. N. Engl. J. Med. 2018, 378, 11–21. [Google Scholar] [CrossRef]
  9. Amantea, D.; Greco, R. Neuroprotection Following Stroke. In Comprehensive Pharmacology; Elsevier: Amsterdam, The Netherlands, 2022; pp. 64–90. [Google Scholar] [CrossRef]
  10. Haupt, M.; Gerner, S.T.; Bähr, M.; Doeppner, T.R. Quest for Quality in Translational Stroke Research—A New Dawn for Neuroprotection? Int. J. Mol. Sci. 2022, 23, 5381. [Google Scholar] [CrossRef]
  11. Chamorro, Á.; Dirnagl, U.; Urra, X.; Planas, A.M. Neuroprotection in acute stroke: Targeting excitotoxicity, oxidative and nitrosative stress, and inflammation. Lancet Neurol. 2016, 15, 869–881. [Google Scholar] [CrossRef]
  12. Dirnagl, U. Pathobiology of injury after stroke: The neurovascular unit and beyond. Ann. N. Y. Acad. Sci. 2012, 1268, 21–25. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, F.; Xie, X.; Xing, X.; Sun, X. Excitatory Synaptic Transmission in Ischemic Stroke: A New Outlet for Classical Neuroprotective Strategies. Int. J. Mol. Sci. 2022, 23, 9381. [Google Scholar] [CrossRef] [PubMed]
  14. Amantea, D.; Greco, R.; Micieli, G.; Bagetta, G. Paradigm Shift to Neuroimmunomodulation for Translational Neuroprotection in Stroke. Front. Neurosci. 2018, 12, 241. [Google Scholar] [CrossRef] [PubMed]
  15. Endres, M.; Moro, M.A.; Nolte, C.H.; Dames, C.; Buckwalter, M.S.; Meisel, A. Immune Pathways in Etiology, Acute Phase, and Chronic Sequelae of Ischemic Stroke. Circ. Res. 2022, 130, 1167–1186. [Google Scholar] [CrossRef]
  16. Frank, D.; Zlotnik, A.; Boyko, M.; Gruenbaum, B.F. The Development of Novel Drug Treatments for Stroke Patients: A Review. Int. J. Mol. Sci. 2022, 23, 5796. [Google Scholar] [CrossRef]
  17. Gülke, E.; Gelderblom, M.; Magnus, T. Danger signals in stroke and their role on microglia activation after ischemia. Ther. Adv. Neurol. Disord. 2018, 11, 1756286418774254. [Google Scholar] [CrossRef] [Green Version]
  18. Kumar, V. Toll-like receptors in the pathogenesis of neuroinflammation. J. Neuroimmunol. 2019, 332, 16–30. [Google Scholar] [CrossRef]
  19. Balança, B.; Desmurs, L.; Grelier, J.; Perret-Liaudet, A.; Lukaszewicz, A.-C. DAMPs and RAGE Pathophysiology at the Acute Phase of Brain Injury: An Overview. Int. J. Mol. Sci. 2021, 22, 2439. [Google Scholar] [CrossRef]
  20. Durán-Laforet, V.; Peña-Martínez, C.; García-Culebras, A.; Alzamora, L.; Moro, M.; Lizasoain, I. Pathophysiological and pharmacological relevance of TLR4 in peripheral immune cells after stroke. Pharmacol. Ther. 2021, 228, 107933. [Google Scholar] [CrossRef]
  21. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef]
  22. Fang, H.; Chen, J.; Lin, S.; Wang, P.; Wang, Y.; Xiong, X.; Yang, Q. CD36-Mediated Hematoma Absorption following Intracerebral Hemorrhage: Negative Regulation by TLR4 Signaling. J. Immunol. 2014, 192, 5984–5992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Li, F.; Faustino, J.; Woo, M.-S.; Derugin, N.; Vexler, Z.S. Lack of the scavenger receptor CD36 alters microglial phenotypes after neonatal stroke. J. Neurochem. 2015, 135, 445–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Schilling, M.; Besselmann, M.; Müller, M.; Strecker, J.K.; Ringelstein, E.B.; Kiefer, R. Predominant phagocytic activity of resident microglia over hematogenous macrophages following transient focal cerebral ischemia: An investigation using green fluorescent protein transgenic bone marrow chimeric mice. Exp. Neurol. 2005, 196, 290–297. [Google Scholar] [CrossRef] [PubMed]
  25. Hu, X.; Li, P.; Guo, Y.; Wang, H.; Leak, R.K.; Chen, S.; Gao, Y.; Chen, J. Microglia/Macrophage Polarization Dynamics Reveal Novel Mechanism of Injury Expansion After Focal Cerebral Ischemia. Stroke 2012, 43, 3063–3070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Amantea, D.; Bagetta, G.; Tassorelli, C.; Mercuri, N.B.; Corasaniti, M.T. Identification of distinct cellular pools of interleukin-1β during the evolution of the neuroinflammatory response induced by transient middle cerebral artery occlusion in the brain of rat. Brain Res. 2010, 1313, 259–269. [Google Scholar] [CrossRef] [PubMed]
  27. Amantea, D.; Tassorelli, C.; Petrelli, F.; Certo, M.; Bezzi, P.; Micieli, G.; Corasaniti, M.; Bagetta, G. Understanding the multifaceted role of inflammatory mediators in ischemic stroke. Curr. Med. Chem. 2014, 21, 2098–2117. [Google Scholar] [CrossRef]
  28. Clausen, B.H.; Lambertsen, K.L.; Babcock, A.A.; Holm, T.H.; Dagnaes-Hansen, F.; Finsen, B. Interleukin-1beta and tumor necrosis factor-alpha are expressed by different subsets of microglia and macrophages after ischemic stroke in mice. J. Neuroinflammation 2008, 5, 46. [Google Scholar] [CrossRef] [Green Version]
  29. Fumagalli, S.; Perego, C.; Pischiutta, F.; Zanier, E.; de Simoni, M.G. The Ischemic Environment Drives Microglia and Macrophage Function. Front. Neurol. 2015, 6, 81. [Google Scholar] [CrossRef] [Green Version]
  30. Benakis, C.; Garcia-Bonilla, L.; Iadecola, C.; Anrather, J. The role of microglia and myeloid immune cells in acute cerebral ischemia. Front. Cell. Neurosci. 2015, 8, 461. [Google Scholar] [CrossRef]
  31. Young, A.M.H.; Kumasaka, N.; Calvert, F.; Hammond, T.R.; Knights, A.; Panousis, N.; Park, J.S.; Schwartzentruber, J.; Liu, J.; Kundu, K.; et al. A map of transcriptional heterogeneity and regulatory variation in human microglia. Nat. Genet. 2021, 53, 861–868. [Google Scholar] [CrossRef]
  32. Morris, R.S.; Jones, P.S.; Alawneh, J.; Hong, Y.T.; Fryer, T.D.; Aigbirhio, F.I.; Warburton, E.A.; Baron, J.-C. Relationships between selective neuronal loss and microglial activation after ischaemic stroke in man. Brain 2018, 141, 2098–2111. [Google Scholar] [CrossRef] [Green Version]
  33. Iadecola, C.; Buckwalter, M.S.; Anrather, J. Immune responses to stroke: Mechanisms, modulation, and therapeutic potential. J. Clin. Investig. 2020, 130, 2777–2788. [Google Scholar] [CrossRef] [PubMed]
  34. Hu, X.; Leak, R.K.; Shi, Y.; Suenaga, J.; Gao, Y.; Zheng, P.; Chen, J. Microglial and macrophage polarization—New prospects for brain repair. Nat. Rev. Neurol. 2015, 11, 56–64. [Google Scholar] [CrossRef] [PubMed]
  35. Perego, C.; Fumagalli, S.; De Simoni, M.-G. Temporal pattern of expression and colocalization of microglia/macrophage phenotype markers following brain ischemic injury in mice. J. Neuroinflamm. 2011, 8, 174. [Google Scholar] [CrossRef] [Green Version]
  36. Chu, H.X.; Kim, H.A.; Lee, S.; Moore, J.P.; Chan, C.T.; Vinh, A.; Gelderblom, M.; Arumugam, T.; Broughton, B.R.S.; Drummond, G.; et al. Immune Cell Infiltration in Malignant Middle Cerebral Artery Infarction: Comparison with Transient Cerebral Ischemia. J. Cereb. Blood Flow Metab. 2013, 34, 450–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Gelderblom, M.; Leypoldt, F.; Steinbach, K.; Behrens, D.; Choe, C.-U.; Siler, D.A.; Arumugam, T.V.; Orthey, E.; Gerloff, C.; Tolosa, E.; et al. Temporal and Spatial Dynamics of Cerebral Immune Cell Accumulation in Stroke. Stroke 2009, 40, 1849–1857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Miró-Mur, F.; Urra, X.; Ruiz-Jaén, F.; Pedragosa, J.; Chamorro, A.; Planas, A.M. Antigen-Dependent T Cell Response to Neural Peptides After Human Ischemic Stroke. Front. Cell. Neurosci. 2020, 14, 206. [Google Scholar] [CrossRef]
  39. Planas, A.M. Role of Immune Cells Migrating to the Ischemic Brain. Stroke 2018, 49, 2261–2267. [Google Scholar] [CrossRef]
  40. Ritzel, R.; Patel, A.R.; Grenier, J.M.; Crapser, J.; Verma, R.; Jellison, E.R.; McCullough, L.D. Functional differences between microglia and monocytes after ischemic stroke. J. Neuroinflamm. 2015, 12, 106. [Google Scholar] [CrossRef]
  41. Wang, Y.; Cheng, Y.; Song, Q.; Wei, C.; Liu, J.; Wu, B.; Liu, M. The association between monocyte to high-density lipoprotein ratio and hemorrhagic transformation in patients with acute ischemic stroke. Aging 2020, 12, 2498–2506. [Google Scholar] [CrossRef]
  42. Narasimhan, P.B.; Marcovecchio, P.; Hamers, A.A.; Hedrick, C.C. Nonclassical Monocytes in Health and Disease. Annu. Rev. Immunol. 2019, 37, 439–456. [Google Scholar] [CrossRef] [PubMed]
  43. Boyette, L.B.; Macedo, C.; Hadi, K.; Elinoff, B.D.; Walters, J.T.; Ramaswami, B.; Chalasani, G.; Taboas, J.M.; Lakkis, F.G.; Metes, D.M. Phenotype, function, and differentiation potential of human monocyte subsets. PLoS ONE 2017, 12, e0176460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Urra, X.; Villamor, N.; Amaro, S.; Gomez-Choco, M.; Obach, V.; Oleaga, L.; Planas, A.M.; Chamorro, A. Monocyte Subtypes Predict Clinical Course and Prognosis in Human Stroke. J. Cereb. Blood Flow Metab. 2009, 29, 994–1002. [Google Scholar] [CrossRef] [Green Version]
  45. Yang, Q.-W.; Li, J.-C.; Lu, F.-L.; Ai-Qing, W.; Xiang, J.; Zhang, L.-L.; Huang, Z.-Y.; Wang, J.-Z. Upregulated Expression of Toll-Like Receptor 4 in Monocytes Correlates with Severity of Acute Cerebral Infarction. J. Cereb. Blood Flow Metab. 2008, 28, 1588–1596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kaito, M.; Araya, S.-I.; Gondo, Y.; Fujita, M.; Minato, N.; Nakanishi, M.; Matsui, M. Relevance of Distinct Monocyte Subsets to Clinical Course of Ischemic Stroke Patients. PLoS ONE 2013, 8, e69409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Nadareishvili, Z.; Luby, M.; Leigh, R.; Shah, J.; Lynch, J.K.; Hsia, A.W.; Benson, R.T.; Latour, L.L. An MRI Hyperintense Acute Reperfusion Marker Is Related to Elevated Peripheral Monocyte Count in Acute Ischemic Stroke. J. Neuroimaging 2017, 28, 57–60. [Google Scholar] [CrossRef]
  48. Liberale, L.; Montecucco, F.; Bonaventura, A.; Casetta, I.; Seraceni, S.; Trentini, A.; Padroni, M.; Dallegri, F.; Fainardi, E.; Carbone, F. Monocyte count at onset predicts poststroke outcomes during a 90-day follow-up. Eur. J. Clin. Investig. 2017, 47, 702–710. [Google Scholar] [CrossRef]
  49. Ren, H.; Han, L.; Liu, H.; Wang, L.; Liu, X.; Gao, Y. Decreased Lymphocyte-to-Monocyte Ratio Predicts Poor Prognosis of Acute Ischemic Stroke Treated with Thrombolysis. J. Pharmacol. Exp. Ther. 2017, 23, 5826–5833. [Google Scholar] [CrossRef] [Green Version]
  50. Greco, R.; Demartini, C.; Zanaboni, A.; Tumelero, E.; Elisa, C.; Persico, A.; Morotti, A.; Amantea, D.; Tassorelli, C. Characterization of CB2 Receptor Expression in Peripheral Blood Monocytes of Acute Ischemic Stroke Patients. Transl. Stroke Res. 2020, 12, 550–558. [Google Scholar] [CrossRef]
  51. Zrzavy, T.; Machado-Santos, J.; Christine, S.; Baumgartner, C.; Weiner, H.L.; Butovsky, O.; Lassmann, H. Dominant role of microglial and macrophage innate immune responses in human ischemic infarcts. Brain Pathol. 2017, 28, 791–805. [Google Scholar] [CrossRef]
  52. Chiba, T.; Umegaki, K. Pivotal Roles of Monocytes/Macrophages in Stroke. Mediat. Inflamm. 2013, 2013, 759103. [Google Scholar] [CrossRef] [Green Version]
  53. Petry, K.G.; Boiziau, C.; Dousset, V.; Brochet, B. Magnetic resonance imaging of human brain macrophage infiltration. Neurotherapeutics 2007, 4, 434–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Miró-Mur, F.; Pérez-De-Puig, I.; Ferrer-Ferrer, M.; Urra, X.; Justicia, C.; Chamorro, A.; Planas, A.M. Immature monocytes recruited to the ischemic mouse brain differentiate into macrophages with features of alternative activation. Brain Behav. Immun. 2016, 53, 18–33. [Google Scholar] [CrossRef] [Green Version]
  55. Greco, R.; Demartini, C.; Zanaboni, A.M.; Blandini, F.; Amantea, D.; Tassorelli, C. Endothelial nitric oxide synthase inhibition triggers inflammatory responses in the brain of male rats exposed to ischemia-reperfusion injury. J. Neurosci. Res. 2017, 96, 151–159. [Google Scholar] [CrossRef] [PubMed]
  56. Kronenberg, G.; Uhlemann, R.; Richter, N.; Klempin, F.; Wegner, S.; Staerck, L.; Wolf, S.; Uckert, W.; Kettenmann, H.; Endres, M.; et al. Distinguishing features of microglia- and monocyte-derived macrophages after stroke. Acta Neuropathol. 2017, 135, 551–568. [Google Scholar] [CrossRef] [PubMed]
  57. Wattananit, S.; Tornero, D.; Graubardt, N.; Memanishvili, T.; Monni, E.; Tatarishvili, J.; Miskinyte, G.; Ge, R.; Ahlenius, H.; Lindvall, O.; et al. Monocyte-Derived Macrophages Contribute to Spontaneous Long-Term Functional Recovery after Stroke in Mice. J. Neurosci. 2016, 36, 4182–4195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Ruhnau, J.; Schulze, J.; Dressel, A.; Vogelgesang, A. Thrombosis, Neuroinflammation, and Poststroke Infection: The Multifaceted Role of Neutrophils in Stroke. J. Immunol. Res. 2017, 2017, 5140679. [Google Scholar] [CrossRef] [Green Version]
  59. Jickling, G.C.; Liu, D.; Ander, B.P.; Stamova, B.; Zhan, X.; Sharp, F.R. Targeting Neutrophils in Ischemic Stroke: Translational Insights from Experimental Studies. J. Cereb. Blood Flow Metab. 2015, 35, 888–901. [Google Scholar] [CrossRef] [Green Version]
  60. Garcia-Culebras, A.; Durán-Laforet, V.; Peña-Martínez, C.; Moraga, A.; Ballesteros, I.; Cuartero, M.; De La Parra, J.; Palma-Tortosa, S.; Hidalgo, A.; Corbí, A.L.; et al. Role of TLR4 (Toll-Like Receptor 4) in N1/N2 Neutrophil Programming After Stroke. Stroke 2019, 50, 2922–2932. [Google Scholar] [CrossRef]
  61. Cuartero, M.I.; Ballesteros, I.; Moraga, A.; Nombela, F.; Vivancos, J.; Hamilton, J.A.; Corbí, L.; Lizasoain, I.; Moro, M.A. N2 Neutrophils, Novel Players in Brain Inflammation After Stroke. Stroke 2013, 44, 3498–3508. [Google Scholar] [CrossRef] [Green Version]
  62. Frieler, R.A.; Chung, Y.; Ahlers, C.G.; Gheordunescu, G.; Song, J.; Vigil, T.M.; Shah, Y.M.; Mortensen, R.M. Genetic neutrophil deficiency ameliorates cerebral ischemia-reperfusion injury. Exp. Neurol. 2017, 298, 104–111. [Google Scholar] [CrossRef] [PubMed]
  63. Kang, L.; Yu, H.; Yang, X.; Zhu, Y.; Bai, X.; Wang, R.; Cao, Y.; Xu, H.; Luo, H.; Lu, L.; et al. Neutrophil extracellular traps released by neutrophils impair revascularization and vascular remodeling after stroke. Nat. Commun. 2020, 11, 2488. [Google Scholar] [CrossRef]
  64. Peña-Martínez, C.; Durán-Laforet, V.; Garcia-Culebras, A.; Ostos, F.; Hernández-Jiménez, M.; Bravo-Ferrer, I.; Pérez-Ruiz, A.; Ballenilla, F.; Díaz-Guzmán, J.; Pradillo, J.; et al. Pharmacological Modulation of Neutrophil Extracellular Traps Reverses Thrombotic Stroke tPA (Tissue-Type Plasminogen Activator) Resistance. Stroke 2019, 50, 3228–3237. [Google Scholar] [CrossRef] [PubMed]
  65. Hermann, D.M.; Kleinschnitz, C.; Gunzer, M. Implications of polymorphonuclear neutrophils for ischemic stroke and intracerebral hemorrhage: Predictive value, pathophysiological consequences and utility as therapeutic target. J. Neuroimmunol. 2018, 321, 138–143. [Google Scholar] [CrossRef] [PubMed]
  66. Barr, T.L.; Vangilder, R.; Rellick, S.; Brooks, S.D.; Doll, D.N.; Lucke-Wold, A.N.; Chen, D.; Denvir, J.; Warach, S.; Singleton, A.; et al. A Genomic Profile of the Immune Response to Stroke with Implications for Stroke Recovery. Biol. Res. Nurs. 2014, 17, 248–256. [Google Scholar] [CrossRef] [PubMed]
  67. Smith, W.S.; Sung, G.; Starkman, S.; Saver, J.; Kidwell, C.S.; Gobin, Y.; Lutsep, H.L.; Nesbit, G.M.; Grobelny, T.; Rymer, M.M.; et al. Safety and Efficacy of Mechanical Embolectomy in Acute Ischemic Stroke. Stroke 2005, 36, 1432–1438. [Google Scholar] [CrossRef]
  68. Cai, W.; Liu, S.; Hu, M.; Huang, F.; Zhu, Q.; Qiu, W.; Hu, X.; Colello, J.; Zheng, S.G.; Lu, Z. Functional Dynamics of Neutrophils After Ischemic Stroke. Transl. Stroke Res. 2019, 11, 108–121. [Google Scholar] [CrossRef]
  69. Wang, X.; Wang, Q.; Wang, K.; Ni, Q.; Li, H.; Su, Z.; Xu, Y. Is Immune Suppression Involved in the Ischemic Stroke? A Study Based on Computational Biology. Front. Aging Neurosci. 2022, 14, 830494. [Google Scholar] [CrossRef]
  70. Lambertsen, K.L.; Finsen, B.; Clausen, B.H. Post-stroke inflammation—Target or tool for therapy? Acta Neuropathol. 2018, 137, 693–714. [Google Scholar] [CrossRef]
  71. Xue, Y.; Zeng, X.; Tu, W.-J.; Zhao, J. Tumor Necrosis Factor-α: The Next Marker of Stroke. Dis. Markers 2022, 2022, 2395269. [Google Scholar] [CrossRef]
  72. Brambilla, R.; Ashbaugh, J.J.; Magliozzi, R.; Dellarole, A.; Karmally, S.; Szymkowski, D.; Bethea, J.R. Inhibition of soluble tumour necrosis factor is therapeutic in experimental autoimmune encephalomyelitis and promotes axon preservation and remyelination. Brain 2011, 134, 2736–2754. [Google Scholar] [CrossRef] [PubMed]
  73. Yli-Karjanmaa, M.; Clausen, B.H.; Degn, M.; Novrup, H.G.; Ellman, D.G.; Toft-Jensen, P.; Szymkowski, D.E.; Stensballe, A.; Meyer, M.; Brambilla, R.; et al. Topical Administration of a Soluble TNF Inhibitor Reduces Infarct Volume After Focal Cerebral Ischemia in Mice. Front. Neurosci. 2019, 13, 781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Taoufik, E.; Tseveleki, V.; Chu, S.Y.; Tselios, T.; Karin, M.; Lassmann, H.; Szymkowski, D.; Probert, L. Transmembrane tumour necrosis factor is neuroprotective and regulates experimental autoimmune encephalomyelitis via neuronal nuclear factor-κB. Brain 2011, 134, 2722–2735. [Google Scholar] [CrossRef]
  75. Madsen, P.; Clausen, B.H.; Degn, M.; Thyssen, S.; Kristensen, L.K.; Svensson, M.; Ditzel, N.; Finsen, B.; Deierborg, T.; Brambilla, R.; et al. Genetic ablation of soluble tumor necrosis factor with preservation of membrane tumor necrosis factor is associated with neuroprotection after focal cerebral ischemia. J. Cereb. Blood Flow Metab. 2016, 36, 1553–1569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Liguz-Lecznar, M.; Zakrzewska, R.; Kossut, M. Inhibition of Tnf-α R1 signaling can rescue functional cortical plasticity impaired in early post-stroke period. Neurobiol. Aging 2015, 36, 2877–2884. [Google Scholar] [CrossRef]
  77. Sairanen, T.; Lindsberg, P.; Brenner, M.; Carpén, O.; Sirén, A.-L. Differential cellular expression of tumor necrosis factor-α and Type I tumor necrosis factor receptor after transient global forebrain ischemia. J. Neurol. Sci. 2001, 186, 87–99. [Google Scholar] [CrossRef]
  78. Lambertsen, K.; Clausen, B.; Fenger, C.; Wulf, H.; Owens, T.; Dagnaes-Hansen, F.; Meldgaard, M.; Finsen, B. Microglia and macrophages express tumor necrosis factor receptor p75 following middle cerebral artery occlusion in mice. Neuroscience 2007, 144, 934–949. [Google Scholar] [CrossRef]
  79. Dziewulska, D.; Mossakowski, M.J. Cellular expression of tumor necrosis factor a and its receptors in human ischemic stroke. Clin. Neuropathol. 2003, 22, 35–40. [Google Scholar]
  80. Clausen, B.H.; Wirenfeldt, M.; Høgedal, S.S.; Frich, L.H.; Nielsen, H.H.; Schrøder, H.D.; Østergaard, K.; Finsen, B.; Kristensen, B.W.; Lambertsen, K.L. Characterization of the TNF and IL-1 systems in human brain and blood after ischemic stroke. Acta Neuropathol. Commun. 2020, 8, 1–17. [Google Scholar] [CrossRef]
  81. Greco, R.; Demartini, C.; Zanaboni, A.M.; Blandini, F.; Amantea, D.; Tassorelli, C. Modulation of cerebral RAGE expression following nitric oxide synthase inhibition in rats subjected to focal cerebral ischemia. Eur. J. Pharmacol. 2017, 800, 16–22. [Google Scholar] [CrossRef]
  82. Gregersen, R.; Lambertsen, K.; Finsen, B. Microglia and Macrophages Are the Major Source of Tumor Necrosis Factor in Permanent Middle Cerebral Artery Occlusion in Mice. J. Cereb. Blood Flow Metab. 2000, 20, 53–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Greco, R.; Demartini, C.; Zanaboni, A.; Tumelero, E.; Persico, A.; Candeloro, E.; Morotti, A.; Amantea, D.; Tassorelli, C. CD163 as a Potential Biomarker of Monocyte Activation in Ischemic Stroke Patients. Int. J. Mol. Sci. 2021, 22, 6712. [Google Scholar] [CrossRef] [PubMed]
  84. Hansen, R.; Laursen, C.; Nawaz, N.; Madsen, J.; Nielsen, H.; Kruuse, C.; Møller, A.; Degn, M.; Lambertsen, K. Leukocyte TNFR1 and TNFR2 Expression Contributes to the Peripheral Immune Response in Cases with Ischemic Stroke. Cells 2021, 10, 861. [Google Scholar] [CrossRef] [PubMed]
  85. Lee, T.H.; Wisniewski, H.G.; Vilcek, J. A novel secretory tumor necrosis factor-inducible protein (TSG-6) is a member of the family of hyaluronate binding proteins, closely related to the adhesion receptor CD44. J. Cell Biol. 1992, 116, 545–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lee, T.H.; Lee, G.W.; Ziff, E.B.; Vilcek, J. Isolation and characterization of eight tumor necrosis factor-induced gene sequences from human fibroblasts. Mol. Cell. Biol. 1990, 10, 1982–1988. [Google Scholar] [CrossRef]
  87. Wisniewski, H.G.; Maier, R.; Lotz, M.; Lee, S.; Klampfer, L.; Lee, T.H.; Vilcek, J. TSG-6: A TNF-, IL-1-, and LPS-inducible secreted glycoprotein associated with arthritis. J. Immunol. 1993, 151, 6593–6601. [Google Scholar] [CrossRef]
  88. Klampfer, L.; Lee, T.H.; Hsu, W.; Vilcek, J.; Chen-Kiang, S. NF-IL6 and AP-1 cooperatively modulate the activation of the TSG-6 gene by tumor necrosis factor alpha and interleukin-1. Mol. Cell. Biol. 1994, 14, 6561–6569. [Google Scholar] [CrossRef]
  89. Bogdani, M.; Johnson, P.Y.; Potter-Perigo, S.; Nagy, N.; Day, A.J.; Bollyky, P.L.; Wight, T.N. Hyaluronan and Hyaluronan-Binding Proteins Accumulate in Both Human Type 1 Diabetic Islets and Lymphoid Tissues and Associate with Inflammatory Cells in Insulitis. Diabetes 2014, 63, 2727–2743. [Google Scholar] [CrossRef] [Green Version]
  90. Tan, K.T.; McGrouther, D.A.; Day, A.; Milner, C.M.; Bayat, A. Characterization of hyaluronan and TSG-6 in skin scarring: Differential distribution in keloid scars, normal scars and unscarred skin. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 317–327. [Google Scholar] [CrossRef]
  91. Zhang, S.; He, H.; Day, A.J.; Tseng, S.C.G. Constitutive Expression of Inter-α-inhibitor (IαI) Family Proteins and Tumor Necrosis Factor-stimulated Gene-6 (TSG-6) by Human Amniotic Membrane Epithelial and Stromal Cells Supporting Formation of the Heavy Chain-Hyaluronan (HC-HA) Complex. J. Biol. Chem. 2012, 287, 12433–12444. [Google Scholar] [CrossRef]
  92. Coulson-Thomas, V.J.; Lauer, M.E.; Soleman, S.; Zhao, C.; Hascall, V.C.; Day, A.J.; Fawcett, J.W. Tumor Necrosis Factor-stimulated Gene-6 (TSG-6) Is Constitutively Expressed in Adult Central Nervous System (CNS) and Associated with Astrocyte-mediated Glial Scar Formation following Spinal Cord Injury. J. Biol. Chem. 2016, 291, 19939–19952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Milner, C.M.; Day, A.J. TSG-6: A multifunctional protein associated with inflammation. J. Cell Sci. 2003, 116, 1863–1873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Day, A.J.; Milner, C.M. TSG-6: A multifunctional protein with anti-inflammatory and tissue-protective properties. Matrix Biol. 2018, 78–79, 60–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lesley, J.; Gál, I.; Mahoney, D.J.; Cordell, M.R.; Rugg, M.S.; Hyman, R.; Day, A.; Mikecz, K. TSG-6 Modulates the Interaction between Hyaluronan and Cell Surface CD44. J. Biol. Chem. 2004, 279, 25745–25754. [Google Scholar] [CrossRef] [Green Version]
  96. Kota, D.J.; Wiggins, L.L.; Yoon, N.; Lee, R.H. TSG-6 Produced by hMSCs Delays the Onset of Autoimmune Diabetes by Suppressing Th1 Development and Enhancing Tolerogenicity. Diabetes 2013, 62, 2048–2058. [Google Scholar] [CrossRef] [Green Version]
  97. Baranova, N.; Nilebäck, E.; Haller, F.M.; Briggs, D.; Svedhem, S.; Day, A.J.; Richter, R.P. The Inflammation-associated Protein TSG-6 Cross-links Hyaluronan via Hyaluronan-induced TSG-6 Oligomers. J. Biol. Chem. 2011, 286, 25675–25686. [Google Scholar] [CrossRef] [Green Version]
  98. Choi, H.; Lee, R.H.; Bazhanov, N.; Oh, J.Y.; Prockop, D.J. Anti-inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-induced mouse peritonitis by decreasing TLR2/NF-κB signaling in resident macrophages. Blood 2011, 118, 330–338. [Google Scholar] [CrossRef] [Green Version]
  99. Baranova, N.S.; Inforzato, A.; Briggs, D.C.; Tilakaratna, V.; Enghild, J.J.; Thakar, D.; Milner, C.M.; Day, A.J.; Richter, R.P. Incorporation of Pentraxin 3 into Hyaluronan Matrices is Tightly Regulated and Promotes Matrix Cross-linking. J. Biol. Chem. 2014, 289, 30481–30498. [Google Scholar] [CrossRef] [Green Version]
  100. Baranova, N.; Foulcer, S.J.; Briggs, D.; Tilakaratna, V.; Enghild, J.J.; Milner, C.; Day, A.J.; Richter, R.P. Inter-α-inhibitor Impairs TSG-6-induced Hyaluronan Cross-linking. J. Biol. Chem. 2013, 288, 29642–29653. [Google Scholar] [CrossRef] [Green Version]
  101. Coulson-Thomas, V.J.; Gesteira, T.F.; Hascall, V.; Kao, W. Umbilical Cord Mesenchymal Stem Cells Suppress Host Rejection. J. Biol. Chem. 2014, 289, 23465–23481. [Google Scholar] [CrossRef]
  102. Lauer, M.E.; Loftis, J.; de la Motte, C.; Hascall, V.C. Analysis of the Heavy-Chain Modification and TSG-6 Activity in Pathological Hyaluronan Matrices. In Glycosaminoglycans; Humana Press: New York, NY, USA, 2015; pp. 543–548. [Google Scholar] [CrossRef] [Green Version]
  103. Magaña-Guerrero, F.S.; Domínguez-López, A.; Martínez-Aboytes, P.; Buentello-Volante, B.; Garfias, Y. Human Amniotic Membrane Mesenchymal Stem Cells inhibit Neutrophil Extracellular Traps through TSG-6. Sci. Rep. 2017, 7, 12426. [Google Scholar] [CrossRef] [PubMed]
  104. Lee, R.H.; Pulin, A.A.; Seo, M.J.; Kota, D.J.; Ylostalo, J.; Larson, B.L.; Semprun-Prieto, L.; Delafontaine, P.; Prockop, D.J. Intravenous hMSCs Improve Myocardial Infarction in Mice because Cells Embolized in Lung Are Activated to Secrete the Anti-inflammatory Protein TSG-6. Cell Stem Cell 2009, 5, 54–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Oh, J.Y.; Roddy, G.W.; Choi, H.; Lee, R.H.; Ylöstalo, J.H.; Rosa, R.H.; Prockop, D.J. Anti-inflammatory protein TSG-6 reduces inflammatory damage to the cornea following chemical and mechanical injury. Proc. Natl. Acad. Sci. USA 2010, 107, 16875–16880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Watanabe, R.; Sato, Y.; Ozawa, N.; Takahashi, Y.; Koba, S.; Watanabe, T. Emerging Roles of Tumor Necrosis Factor-Stimulated Gene-6 in the Pathophysiology and Treatment of Atherosclerosis. Int. J. Mol. Sci. 2018, 19, 465. [Google Scholar] [CrossRef] [Green Version]
  107. Mittal, M.; Tiruppathi, C.; Nepal, S.; Zhao, Y.-Y.; Grzych, D.; Soni, D.; Prockop, D.J.; Malik, A.B. TNFα-stimulated gene-6 (TSG6) activates macrophage phenotype transition to prevent inflammatory lung injury. Proc. Natl. Acad. Sci. USA 2016, 113, E8151–E8158. [Google Scholar] [CrossRef] [Green Version]
  108. Hu, X.; Liu, L.; Wang, Y.; Yu, Y.; Li, Z.; Liu, Y.; Chai, J. Human Umbilical Cord-Derived Mesenchymal Stem Cells Alleviate Acute Lung Injury Caused by Severe Burn via Secreting TSG-6 and Inhibiting Inflammatory Response. Stem Cells Int. 2022, 2022, 8661689. [Google Scholar] [CrossRef]
  109. Hu, T.; Liu, Y.; Li, X.; Li, X.; Liu, Y.; Wang, Q.; Huang, J.; Yu, J.; Wu, Y.; Chen, S.; et al. Tumor necrosis factor-alpha stimulated gene-6: A biomarker reflecting disease activity in rheumatoid arthritis. J. Clin. Lab. Anal. 2022, 36, e24395. [Google Scholar] [CrossRef]
  110. Mindrescu, C.; Dias, A.A.M.; Olszewski, R.J.; Klein, M.J.; Reis, L.F.L.; Wisniewski, H.-G. Reduced susceptibility to collagen-induced arthritis in DBA/1J mice expressing the TSG-6 transgene. Arthritis Care Res. 2002, 46, 2453–2464. [Google Scholar] [CrossRef]
  111. Mindrescu, C.; Thorbecke, G.J.; Klein, M.J.; Vilček, J.; Wisniewski, H.-G. Amelioration of collagen-induced arthritis in DBA/1J mice by recombinant TSG-6, a tumor necrosis factor/interleukin-1-inducible protein. Arthritis Care Res. 2000, 43, 2668–2677. [Google Scholar] [CrossRef]
  112. Balog, A.; Varga, B.; Fülöp, F.; Lantos, I.; Toldi, G.; Vécsei, L.; Mándi, Y. Kynurenic Acid Analog Attenuates the Production of Tumor Necrosis Factor-α, Calgranulins (S100A 8/9 and S100A 12), and the Secretion of HNP1–3 and Stimulates the Production of Tumor Necrosis Factor-Stimulated Gene-6 in Whole Blood Cultures of Patients with Rheumatoid Arthritis. Front. Immunol. 2021, 12, 632513. [Google Scholar] [CrossRef]
  113. Bárdos, T.; Kamath, R.V.; Mikecz, K.; Glant, T.T. Anti-Inflammatory and Chondroprotective Effect of TSG-6 (Tumor Necrosis Factor-α-Stimulated Gene-6) in Murine Models of Experimental Arthritis. Am. J. Pathol. 2001, 159, 1711–1721. [Google Scholar] [CrossRef] [PubMed]
  114. Liao, Z.; Wang, W.; Deng, W.; Zhang, Y.; Song, A.; Deng, S.; Zhao, H.; Zhang, S.; Li, Z. Human Umbilical Cord Mesenchymal Stem Cells-Secreted TSG-6 Is Anti-Inflammatory and Promote Tissue Repair After Spinal Cord Injury. ASN Neuro 2021, 13, 175909142110106. [Google Scholar] [CrossRef]
  115. Tang, B.; Song, M.; Xie, X.; Le, D.; Tu, Q.; Wu, X.; Chen, M. Tumor Necrosis Factor-stimulated Gene-6 (TSG-6) Secreted by BMSCs Regulates Activated Astrocytes by Inhibiting NF-κB Signaling Pathway to Ameliorate Blood Brain Barrier Damage After Intracerebral Hemorrhage. Neurochem. Res. 2021, 46, 2387–2402. [Google Scholar] [CrossRef] [PubMed]
  116. Li, R.; Liu, W.; Yin, J.; Chen, Y.; Guo, S.; Fan, H.; Li, X.; Zhang, X.; He, X.; Duan, C. TSG-6 attenuates inflammation-induced brain injury via modulation of microglial polarization in SAH rats through the SOCS3/STAT3 pathway. J. Neuroinflamm. 2018, 15, 231. [Google Scholar] [CrossRef] [Green Version]
  117. Watanabe, J.; Shetty, A.K.; Hattiangady, B.; Kim, D.-K.; Foraker, J.E.; Nishida, H.; Prockop, D.J. Administration of TSG-6 improves memory after traumatic brain injury in mice. Neurobiol. Dis. 2013, 59, 86–99. [Google Scholar] [CrossRef] [Green Version]
  118. Roura, S.; Monguió-Tortajada, M.; Munizaga-Larroudé, M.; Clos-Sansalvador, M.; Franquesa, M.; Rosell, A.; Borràs, F.E. Potential of Extracellular Vesicle-Associated TSG-6 from Adipose Mesenchymal Stromal Cells in Traumatic Brain Injury. Int. J. Mol. Sci. 2020, 21, 6761. [Google Scholar] [CrossRef]
  119. Lin, Q.-M.; Zhao, S.; Zhou, L.-L.; Fang, X.-S.; Fu, Y.; Huang, Z.-T. Mesenchymal stem cells transplantation suppresses inflammatory responses in global cerebral ischemia: Contribution of TNF-α-induced protein 6. Acta Pharmacol. Sin. 2013, 34, 784–792. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, R.; Liu, Y.; Yan, K.; Chen, L.; Chen, X.-R.; Li, P.; Chen, F.-F.; Jiang, X.-D. Anti-inflammatory and immunomodulatory mechanisms of mesenchymal stem cell transplantation in experimental traumatic brain injury. J. Neuroinflamm. 2013, 10, 871. [Google Scholar] [CrossRef] [Green Version]
  121. Dyer, D.P.; Thomson, J.M.; Hermant, A.; Jowitt, T.A.; Handel, T.M.; Proudfoot, A.E.I.; Day, A.J.; Milner, C.M. TSG-6 Inhibits Neutrophil Migration via Direct Interaction with the Chemokine CXCL8. J. Immunol. 2014, 192, 2177–2185. [Google Scholar] [CrossRef]
  122. Dyer, D.P.; Salanga, C.L.; Johns, S.C.; Valdambrini, E.; Fuster, M.M.; Milner, C.M.; Day, A.J.; Handel, T.M. The Anti-inflammatory Protein TSG-6 Regulates Chemokine Function by Inhibiting Chemokine/Glycosaminoglycan Interactions. J. Biol. Chem. 2016, 291, 12627–12640. [Google Scholar] [CrossRef] [Green Version]
  123. Getting, S.J.; Mahoney, D.J.; Cao, T.; Rugg, M.S.; Fries, E.; Milner, C.; Perretti, M.; Day, A. The Link Module from Human TSG-6 Inhibits Neutrophil Migration in a Hyaluronan- and Inter-α-inhibitor-independent Manner. J. Biol. Chem. 2002, 277, 51068–51076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wisniewski, H.G.; Hua, J.C.; Poppers, D.M.; Naime, D.; Vilcek, J.; Cronstein, B.N. TNF/IL-1-inducible protein TSG-6 potentiates plasmin inhibition by inter-alpha-inhibitor and exerts a strong anti-inflammatory effect in vivo. J. Immunol. 1996, 156, 1609–1615. [Google Scholar] [CrossRef] [PubMed]
  125. Cao, T.V.; La, M.; Getting, S.J.; Day, A.J.; Perretti, M. Inhibitory Effects of TSG-6 Link Module on Leukocyte–Endothelial Cell Interactions In Vitro and In Vivo. Microcirculation 2004, 11, 615–624. [Google Scholar] [CrossRef] [PubMed]
  126. Hertsenberg, A.J.; Shojaati, G.; Funderburgh, M.L.; Mann, M.M.; Du, Y.; Funderburgh, J.L. Corneal stromal stem cells reduce corneal scarring by mediating neutrophil infiltration after wounding. PLoS ONE 2017, 12, e0171712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Liu, L.; Yu, Y.; Hou, Y.; Chai, J.; Duan, H.; Chu, W.; Zhang, H.; Hu, Q.; Du, J. Human Umbilical Cord Mesenchymal Stem Cells Transplantation Promotes Cutaneous Wound Healing of Severe Burned Rats. PLoS ONE 2014, 9, e88348. [Google Scholar] [CrossRef] [Green Version]
  128. Szántó, S.; Bárdos, T.; Gál, I.; Glant, T.T.; Mikecz, K. Enhanced neutrophil extravasation and rapid progression of proteoglycan-induced arthritis in TSG-6-knockout mice. Arthritis Care Res. 2004, 50, 3012–3022. [Google Scholar] [CrossRef] [Green Version]
  129. Qi, Y.; Jiang, D.; Sindrilaru, A.; Stegemann, A.; Schatz, S.; Treiber, N.; Rojewski, M.; Schrezenmeier, H.; Vander Beken, S.; Wlaschek, M.; et al. TSG-6 Released from Intradermally Injected Mesenchymal Stem Cells Accelerates Wound Healing and Reduces Tissue Fibrosis in Murine Full-Thickness Skin Wounds. J. Investig. Dermatol. 2014, 134, 526–537. [Google Scholar] [CrossRef] [Green Version]
  130. Kim, S.J.; Lee, H.J.; Yun, J.-H.; Ko, J.H.; Choi, D.Y.; Oh, J.Y. Intravitreal TSG-6 suppresses laser-induced choroidal neovascularization by inhibiting CCR2+ monocyte recruitment. Sci. Rep. 2015, 5, 11872. [Google Scholar] [CrossRef] [Green Version]
  131. Oh, J.Y.; Lee, R.H.; Yu, J.M.; Ko, J.H.; Lee, H.J.; Ko, A.Y.; Roddy, G.W.; Prockop, D.J. Intravenous Mesenchymal Stem Cells Prevented Rejection of Allogeneic Corneal Transplants by Aborting the Early Inflammatory Response. Mol. Ther. 2012, 20, 2143–2152. [Google Scholar] [CrossRef]
  132. Kui, L.; Chan, G.C.F.; Lee, P.P.W. TSG-6 Downregulates IFN-Alpha and TNF-Alpha Expression by Suppressing IRF7 Phosphorylation in Human Plasmacytoid Dendritic Cells. Mediat. Inflamm. 2017, 2017, 7462945. [Google Scholar] [CrossRef] [Green Version]
  133. Li, Y.; Zhang, D.; Xu, L.; Dong, L.; Zheng, J.; Lin, Y.; Huang, J.; Zhang, Y.; Tao, Y.; Zang, X.; et al. Cell–cell contact with proinflammatory macrophages enhances the immunotherapeutic effect of mesenchymal stem cells in two abortion models. Cell. Mol. Immunol. 2019, 16, 908–920. [Google Scholar] [CrossRef] [PubMed]
  134. Maina, V.; Cotena, A.; Doni, A.; Nebuloni, M.; Pasqualini, F.; Milner, C.M.; Day, A.J.; Mantovani, A.; Garlanda, C. Coregulation in human leukocytes of the long pentraxin PTX3 and TSG-6. J. Leukoc. Biol. 2009, 86, 123–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Li, C.; Li, X.; Shi, Z.; Wu, P.; Fu, J.; Tang, J.; Qing, L. Exosomes from LPS-preconditioned bone marrow MSCs accelerated peripheral nerve regeneration via M2 macrophage polarization: Involvement of TSG-6/NF-κB/NLRP3 signaling pathway. Exp. Neurol. 2022, 356, 114139. [Google Scholar] [CrossRef] [PubMed]
  136. Bartosh, T.J.; Ylöstalo, J.H.; Mohammadipoor, A.; Bazhanov, N.; Coble, K.; Claypool, K.; Lee, R.H.; Choi, H.; Prockop, D.J. Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties. Proc. Natl. Acad. Sci. USA 2010, 107, 13724–13729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Greco, R.; Mangione, A.S.; Amantea, D.; Bagetta, G.; Nappi, G.; Tassorelli, C. IkappaB-alpha expression following transient focal cerebral ischemia is modulated by nitric oxide. Brain Res. 2011, 1372, 145–151. [Google Scholar] [CrossRef]
  138. Lee, R.H.; Yu, J.M.; Foskett, A.M.; Peltier, G.; Reneau, J.C.; Bazhanov, N.; Oh, J.Y.; Prockop, D.J. TSG-6 as a biomarker to predict efficacy of human mesenchymal stem/progenitor cells (hMSCs) in modulating sterile inflammation in vivo. Proc. Natl. Acad. Sci. USA 2014, 111, 16766–16771. [Google Scholar] [CrossRef] [Green Version]
  139. Wan, Y.; Song, M.; Xie, X.; Chen, Z.; Gao, Z.; Wu, X.; Huang, R.; Chen, M. BMSCs Regulate Astrocytes through TSG-6 to Protect the Blood-Brain Barrier after Subarachnoid Hemorrhage. Mediat. Inflamm. 2021, 2021, 5522291. [Google Scholar] [CrossRef]
  140. Hu, Y.; Li, G.; Zhang, Y.; Liu, N.; Zhang, P.; Pan, C.; Nie, H.; Li, Q.; Tang, Z. Upregulated TSG-6 Expression in ADSCs Inhibits the BV2 Microglia-Mediated Inflammatory Response. BioMed Res. Int. 2018, 2018, 7239181. [Google Scholar] [CrossRef]
  141. Liu, Y.; Zhang, R.; Yan, K.; Chen, F.; Huang, W.; Lv, B.; Sun, C.; Xu, L.; Li, F.; Jiang, X. Mesenchymal stem cells inhibit lipopolysaccharide-induced inflammatory responses of BV2 microglial cells through TSG-6. J. Neuroinflamm. 2014, 11, 135. [Google Scholar] [CrossRef]
  142. Muto, J.; Yamasaki, K.; Taylor, K.R.; Gallo, R.L. Engagement of CD44 by hyaluronan suppresses TLR4 signaling and the septic response to LPS. Mol. Immunol. 2009, 47, 449–456. [Google Scholar] [CrossRef] [Green Version]
  143. Kawana, H.; Karaki, H.; Higashi, M.; Miyazaki, M.; Hilberg, F.; Kitagawa, M.; Harigaya, K. CD44 Suppresses TLR-Mediated Inflammation. J. Immunol. 2008, 180, 4235–4245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Yang, H.; Wu, L.; Deng, H.; Chen, Y.; Zhou, H.; Liu, M.; Wang, S.; Zheng, L.; Zhu, L.; Lv, X. Anti-inflammatory protein TSG-6 secreted by bone marrow mesenchymal stem cells attenuates neuropathic pain by inhibiting the TLR2/MyD88/NF-κB signaling pathway in spinal microglia. J. Neuroinflamm. 2020, 17, 154. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, L.; Song, H.; Duan, H.; Chai, J.; Yang, J.; Li, X.; Yu, Y.; Zhang, X.; Hu, X.; Xiao, M.; et al. TSG-6 secreted by human umbilical cord-MSCs attenuates severe burn-induced excessive inflammation via inhibiting activations of P38 and JNK signaling. Sci. Rep. 2016, 6, 30121. [Google Scholar] [CrossRef] [Green Version]
  146. Zhang, C.; Tao, Q.; Ge, S.; Zhai, Z.; Wang, H. Tumor necrosis factor alpha-stimulated gene-6 (TSG-6) inhibits the inflammatory response by inhibiting the activation of P38 and JNK signaling pathway and decreases the restenosis of vein grafts in rats. Hear. Vessel. 2017, 32, 1536–1545. [Google Scholar] [CrossRef] [PubMed]
  147. Watanabe, R.; Watanabe, H.; Takahashi, Y.; Kojima, M.; Konii, H.; Watanabe, K.; Shirai, R.; Sato, K.; Matsuyama, T.-A.; Ishibashi-Ueda, H.; et al. Atheroprotective Effects of Tumor Necrosis Factor–Stimulated Gene-6. JACC Basic Transl. Sci. 2016, 1, 494–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Deng, W.; Mandeville, E.; Terasaki, Y.; Li, W.; Holder, J.; Chuang, A.T.; Ning, M.; Arai, K.; Lo, E.H.; Xing, C. Transcriptomic characterization of microglia activation in a rat model of ischemic stroke. J. Cereb. Blood Flow Metab. 2020, 40, S34–S48. [Google Scholar] [CrossRef] [PubMed]
  149. Zhou, X.; Zhang, Y.-N.; Li, F.-F.; Zhang, Z.; Cui, L.-Y.; He, H.-Y.; Yan, X.; He, W.-B.; Sun, H.-S.; Feng, Z.-P.; et al. Neuronal chemokine-like-factor 1 (CKLF1) up-regulation promotes M1 polarization of microglia in rat brain after stroke. Acta Pharmacol. Sin. 2021, 43, 1217–1230. [Google Scholar] [CrossRef]
  150. Gaire, B.P.; Song, M.-R.; Choi, J.W. Sphingosine 1-phosphate receptor subtype 3 (S1P3) contributes to brain injury after transient focal cerebral ischemia via modulating microglial activation and their M1 polarization. J. Neuroinflamm. 2018, 15, 284. [Google Scholar] [CrossRef] [Green Version]
  151. Han, D.; Wang, J.; Wen, L.; Sun, M.; Liu, H.; Gao, Y. Remote Limb Ischemic Postconditioning Protects against Ischemic Stroke via Modulating Microglia/Macrophage Polarization in Mice. J. Immunol. Res. 2021, 2021, 6688053. [Google Scholar] [CrossRef]
  152. Li, Q.; Dai, Z.; Cao, Y.; Wang, L. Caspase-1 inhibition mediates neuroprotection in experimental stroke by polarizing M2 microglia/macrophage and suppressing NF-κB activation. Biochem. Biophys. Res. Commun. 2019, 513, 479–485. [Google Scholar] [CrossRef]
  153. Zhang, T.; Wang, D.; Li, X.; Jiang, Y.; Wang, C.; Zhang, Y.; Kong, Q.; Tian, C.; Dai, Y.; Zhao, W.; et al. Excess salt intake promotes M1 microglia polarization via a p38/MAPK/AR-dependent pathway after cerebral ischemia in mice. Int. Immunopharmacol. 2020, 81, 106176. [Google Scholar] [CrossRef] [PubMed]
  154. Jha, K.A.; Pentecost, M.; Lenin, R.; Gentry, J.; Klaic, L.; Del Mar, N.; Reiner, A.; Yang, C.H.; Pfeffer, L.M.; Sohl, N.; et al. TSG-6 in conditioned media from adipose mesenchymal stem cells protects against visual deficits in mild traumatic brain injury model through neurovascular modulation. Stem Cell Res. Ther. 2019, 10, 1–15. [Google Scholar] [CrossRef] [Green Version]
  155. Liu, Y.; Zeng, R.; Wang, Y.; Huang, W.; Hu, B.; Zhu, G.; Zhang, R.; Li, F.; Han, J.; Li, Y. Mesenchymal stem cells enhance microglia M2 polarization and attenuate neuroinflammation through TSG-6. Brain Res. 2019, 1724, 146422. [Google Scholar] [CrossRef] [PubMed]
  156. Wan, Y.-M.; Wu, H.-M.; Li, Y.-H.; Xu, Z.-Y.; Yang, J.-H.; Liu, C.; He, Y.-F.; Wang, M.-J.; Wu, X.-N.; Zhang, Y. TSG-6 Inhibits Oxidative Stress and Induces M2 Polarization of Hepatic Macrophages in Mice with Alcoholic Hepatitis via Suppression of STAT3 Activation. Front. Pharmacol. 2020, 11, 10. [Google Scholar] [CrossRef] [Green Version]
  157. Lin, Q.; Lin, S.; Lv, Y.; Zhou, L.; Fu, Y.; Fang, X.; Chen, F.; Huang, Z. Suppression of inflammatory damage to the brain after global cerebral ischemia by transplanted mesenchymal stem cells via secretion of TSG-6. Neurol. Asia. 2016, 21, 113–122. [Google Scholar]
  158. Jung, H.-S.; Jeong, S.-Y.; Yang, J.; Kim, S.-D.; Zhang, B.; Yoo, H.S.; Song, S.U.; Jeon, M.-S.; Song, Y.S. Neuroprotective effect of mesenchymal stem cell through complement component 3 downregulation after transient focal cerebral ischemia in mice. Neurosci. Lett. 2016, 633, 227–234. [Google Scholar] [CrossRef] [PubMed]
  159. Chen, M.; Li, X.; Zhang, X.; He, X.; Lai, L.; Liu, Y.; Zhu, G.; Li, W.; Li, H.; Fang, Q.; et al. The inhibitory effect of mesenchymal stem cell on blood–brain barrier disruption following intracerebral hemorrhage in rats: Contribution of TSG-6. J. Neuroinflamm. 2015, 12, 61. [Google Scholar] [CrossRef] [Green Version]
  160. Chen, T.; Zhu, J.; Hang, C.-H.; Wang, Y.-H. The Potassium SK Channel Activator NS309 Protects Against Experimental Traumatic Brain Injury Through Anti-Inflammatory and Immunomodulatory Mechanisms. Front. Pharmacol. 2019, 10, 1432. [Google Scholar] [CrossRef]
  161. Bertling, F.; Bendix, I.; Drommelschmidt, K.; Wisniewski, H.; Felderhoff-Mueser, U.; Keller, M.; Prager, S. Tumor necrosis factor-inducible gene 6 protein: A novel neuroprotective factor against inflammation-induced developmental brain injury. Exp. Neurol. 2016, 279, 283–289. [Google Scholar] [CrossRef] [Green Version]
  162. Qteishat, A.A.; Gaffney, J.; Krupinski, J.; Rubio, F.; West, D.; Kumar, S.; Kumar, P.; Mitsios, N.; Slevin, M. Changes in hyaluronan production and metabolism following ischaemic stroke in man. Brain 2006, 129, 2158–2176. [Google Scholar] [CrossRef]
  163. Wang, H.; Zhan, Y.; Xu, L.; Feuerstein, G.Z.; Wang, X. Use of Suppression Subtractive Hybridization for Differential Gene Expression in Stroke. Stroke 2001, 32, 1020–1027. [Google Scholar] [CrossRef] [Green Version]
  164. Wang, X.; Xu, L.; Wang, H.; Zhan, Y.; Puré, E.; Feuerstein, G.Z. CD44 deficiency in mice protects brain from cerebral ischemia injury. J. Neurochem. 2002, 83, 1172–1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Qu, Y.; Yang, F.; Meng, F.; Chen, X.; Zhang, Q.; Yu, T.; Wen, S.; Pan, Y. Plasma Concentration of Tumor Necrosis Factor-Stimulated Gene-6 as a Novel Diagnostic and 3-Month Prognostic Indicator in Non-Cardioembolic Acute Ischemic Stroke. Front. Immunol. 2022, 13, 713379. [Google Scholar] [CrossRef] [PubMed]
  166. Tian, X.; Wang, X.; Shi, Z.; Yu, C.; Li, M.; Chen, L.; Jia, Q.; Liang, G. Tumor necrosis factor-stimulated gene-6-a new serum identification marker to identify severe and symptomatic carotid artery stenosis. Pathol. Res. Pract. 2022, 232, 153838. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, S.K.; Xie, J.; Green, L.A.; McCready, R.A.; Motaganahalli, R.L.; Fajardo, A.; Babbey, C.C.; Murphy, M.P. TSG-6 is highly expressed in human abdominal aortic aneurysms. J. Surg. Res. 2017, 220, 311–319. [Google Scholar] [CrossRef]
  168. Ye, L.; Mora, R.; Akhayani, N.; Haudenschild, C.C.; Liau, G. Growth Factor and Cytokine-Regulated Hyaluronan-Binding Protein TSG-6 Is Localized to the Injury-Induced Rat Neointima and Confers Enhanced Growth in Vascular Smooth Muscle Cells. Circ. Res. 1997, 81, 289–296. [Google Scholar] [CrossRef]
  169. Wang, S.-S.; Hu, S.-W.; Zhang, Q.-H.; Xia, A.-X.; Jiang, Z.-X.; Chen, X.-M. Mesenchymal Stem Cells Stabilize Atherosclerotic Vulnerable Plaque by Anti-Inflammatory Properties. PLoS ONE 2015, 10, e0136026. [Google Scholar] [CrossRef] [Green Version]
  170. Damodarasamy, M.; Vernon, R.B.; Pathan, J.L.; Keene, C.D.; Day, A.J.; Banks, W.A.; Reed, M.J. The microvascular extracellular matrix in brains with Alzheimer’s disease neuropathologic change (ADNC) and cerebral amyloid angiopathy (CAA). Fluids Barriers CNS 2020, 17, 1–11. [Google Scholar] [CrossRef]
  171. Reed, M.J.; Damodarasamy, M.; Pathan, J.L.; Chan, C.K.; Spiekerman, C.; Wight, T.N.; Banks, W.A.; Day, A.J.; Vernon, R.B.; Keene, C.D. Increased Hyaluronan and TSG-6 in Association with Neuropathologic Changes of Alzheimer’s Disease. J. Alzheimer’s Dis. 2019, 67, 91–102. [Google Scholar] [CrossRef]
  172. Mutoji, K.N.; Sun, M.; Nash, A.; Puri, S.; Hascall, V.; Coulson-Thomas, V.J. Anti-inflammatory protein TNFα-stimulated gene-6 (TSG-6) reduces inflammatory response after brain injury in mice. BMC Immunol. 2021, 22, 52. [Google Scholar] [CrossRef]
  173. Li, X.; Liu, W.; Li, R.; Guo, S.; Fan, H.; Wei, B.; Zhang, X.; He, X.; Duan, C. TSG-6 Attenuates Oxidative Stress-Induced Early Brain Injury in Subarachnoid Hemorrhage Partly by the HO-1 and Nox2 Pathways. J. Stroke Cerebrovasc. Dis. 2020, 29, 104986. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Main mechanisms involved in the immunomodulatory functions of TSG-6. Abbreviations: DC: dendritic cells, HA: hyaluronate, HS: heparan sulphate, IL: interleukin, LPS: lipopolysaccharide, MSCs: mesenchymal stem/stromal cells, myeloid differentiation primary response: MyD, TLRs: Toll-like receptors, NF-κB: nuclear factor kappa B, TNF: tumor necrosis factor, TSG: tumor necrosis factor-α-stimulated gene, VSMCs: vascular smooth muscle cells.
Figure 1. Main mechanisms involved in the immunomodulatory functions of TSG-6. Abbreviations: DC: dendritic cells, HA: hyaluronate, HS: heparan sulphate, IL: interleukin, LPS: lipopolysaccharide, MSCs: mesenchymal stem/stromal cells, myeloid differentiation primary response: MyD, TLRs: Toll-like receptors, NF-κB: nuclear factor kappa B, TNF: tumor necrosis factor, TSG: tumor necrosis factor-α-stimulated gene, VSMCs: vascular smooth muscle cells.
Ijms 24 01162 g001
Figure 2. Mechanisms underlying the neuroprotective effects of TSG-6 in rodents subjected to acute brain injury. Abbreviations: BBB: blood–brain barrier, IL: interleukin, INF: interferon, MCP: monocyte chemoattractant protein, MIP: macrophage inflammatory protein, MMP: matrix metalloprotease, MSC: mesenchymal stem/stromal cells, TGF: transforming growth factor, TNF: tumor necrosis factor, TSG: tumor necrosis factor-α-stimulated gene.
Figure 2. Mechanisms underlying the neuroprotective effects of TSG-6 in rodents subjected to acute brain injury. Abbreviations: BBB: blood–brain barrier, IL: interleukin, INF: interferon, MCP: monocyte chemoattractant protein, MIP: macrophage inflammatory protein, MMP: matrix metalloprotease, MSC: mesenchymal stem/stromal cells, TGF: transforming growth factor, TNF: tumor necrosis factor, TSG: tumor necrosis factor-α-stimulated gene.
Ijms 24 01162 g002
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

La Russa, D.; Di Santo, C.; Lizasoain, I.; Moraga, A.; Bagetta, G.; Amantea, D. Tumor Necrosis Factor (TNF)-α-Stimulated Gene 6 (TSG-6): A Promising Immunomodulatory Target in Acute Neurodegenerative Diseases. Int. J. Mol. Sci. 2023, 24, 1162. https://doi.org/10.3390/ijms24021162

AMA Style

La Russa D, Di Santo C, Lizasoain I, Moraga A, Bagetta G, Amantea D. Tumor Necrosis Factor (TNF)-α-Stimulated Gene 6 (TSG-6): A Promising Immunomodulatory Target in Acute Neurodegenerative Diseases. International Journal of Molecular Sciences. 2023; 24(2):1162. https://doi.org/10.3390/ijms24021162

Chicago/Turabian Style

La Russa, Daniele, Chiara Di Santo, Ignacio Lizasoain, Ana Moraga, Giacinto Bagetta, and Diana Amantea. 2023. "Tumor Necrosis Factor (TNF)-α-Stimulated Gene 6 (TSG-6): A Promising Immunomodulatory Target in Acute Neurodegenerative Diseases" International Journal of Molecular Sciences 24, no. 2: 1162. https://doi.org/10.3390/ijms24021162

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