Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
J Clin Med. 2019 Apr; 8(4): 537.
Published online 2019 Apr 19. doi: 10.3390/jcm8040537
PMCID: PMC6518209
PMID: 31010132

T-Regulatory Cells Confer Increased Myelination and Stem Cell Activity after Stroke-Induced White Matter Injury

Abstract

Stroke-induced hypoxia causes oligodendrocyte death due to inflammation, lack of oxygen and exacerbation of cell death. Bone marrow-derived stem cells (BMSCs) possess an endogenous population of T-regulatory cells (Tregs) which reduce secretion of pro-inflammatory cytokines that lead to secondary cell death. Here, we hypothesize that oligodendrocyte progenitor cells (OPCs) cultured with BMSCs containing their native Treg population show greater cell viability, less pro-inflammatory cytokine secretion and greater myelin production after exposure to oxygen-glucose deprivation and reoxygenation (OGD/R) than OPCs cultured without Tregs. OPCs were cultured and then exposed to OGD/R. BMSCs with or without Tregs were added to the co-culture immediately after ischemia. The Tregs were depleted by running the BMSCs through a column containing a magnetic substrate. Fibroblast growth factor beta (FGF-β) and interleukin 6 (IL-6) ELISAs determined BMSC activity levels. Immunohistochemistry assessed OPC differentiation. OPCs cultured with BMSCs containing their endogenous Tregs showed increased myelin production compared to the BMSCs with depleted Tregs. IL-6 and FGF-β were increased in the group cultured with Tregs. Collectively, these results suggest that BMSCs containing Tregs are more therapeutically active, and that Tregs have beneficial effects on OPCs subjected to ischemia. Tregs play an important role in stem cell therapy and can potentially treat white matter injury post-stroke.

Keywords: bone marrow-derived stem cells, mesenchymal stem cells, T-regulatory cells, white matter injury, stroke, oligodendrocytes, oligodendrocyte precursor cells, oxygen–glucose deprivation, reoxygenation

1. Introduction

Although stroke rates are declining, cerebrovascular accidents present a major obstacle for public health in the United States. Stroke is responsible for approximately 1 in 20 deaths in Americans [1]. Ischemic stroke comprises 87% of all strokes [1]. The loss of blood perfusion to brain tissue creates a hypoxic environment for endothelial cells, neurons and other cells critical for providing support to the neurovascular unit, resulting acutely in necrosis [2,3], and the ensuing secondary cell death plagued with activation of immune cells and pro-inflammatory cytokines, such as tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), interleukin 6 (IL-6) and other elements of the innate and adaptive immune systems [4]. Prolonged inflammation can have detrimental effects on brain tissue and can cause further cell death [5].

Clinical safety and efficacy of stem cell transplantation is still being determined [6]. Bone marrow-derived stem cells (BMSCs) have been widely investigated for their potential use in stroke treatment due to their performance in clinical trials, though lack of a sufficient control and sample size hinders their progression into clinical use [7,8]. Endogenous BMSCs and transplanted BMSCs mobilize and home into the inflamed site after an ischemic episode [9]. Self-renewing BMSCs reside in the bone marrow and have the capacity to differentiate into multiple cell lineages, including fibroblasts, adipocytes, placental cells, skeletal myocytes, chondrocytes and others both in vivo and in vitro [10]. In past studies, researchers have extensively used the markers CD271 and CD146 to identify BMSCs [10]. Additional markers of multipotent BMSCs include platelet-derived growth factor alpha and CD105 [10]. BMSCs have long been of interest since the 1800s, when a German scientist hypothesized that fibroblasts originated in the bone marrow [11]. More recently, they have been investigated for the purpose of tissue renewal and their anti-inflammatory secretome, which could be useful in mitigating inflammation caused by pathologic conditions. Mesenchymal stem cells possess an attractive track record of conferring anti-inflammation [12]. When administered in the acute phase of ischemic injury, stem cell transplantation is thought to attenuate oxidative stress, inflammation and cell death [13]. T-regulatory cells (Tregs), a subtype of T-cell, have recently discovered immunosuppressive properties. Tregs express the biomarkers FoxP3, CD25 and CD4 and have been found to exist as a small subpopulation of BMSCs [14]. The native Treg population, at present about 0.4% of BMSCs, was shown to confer increased cell viability via anti-inflammation in a set of neurons subjected to oxygen-glucose deprivation and reoxygenation (OGD/R). After OGD/R, pro-inflammatory cytokine IL-6 decreased with increasing concentrations of Tregs in co-culture [14]. There was a loss of immunosuppressive function after deletion of the FoxP3 allele [15]. Interestingly, the number of Tregs varied inversely with basic fibroblast growth factor beta (FGF-β), suggesting that increasing the number of Tregs beyond that of the native population within BMSCs may have deleterious effects on the survival and differentiation of the BMSCs [14]. M2 microglia possess neuroprotective abilities and have been shown to facilitate white matter repair [16]. Tregs can mitigate inflammation after ischemia by influencing macrophage polarization toward the more regenerative M2 phenotype [17,18,19]. Indeed, a decrease in Tregs is seen after ischemic stroke, which could play a crucial role in exacerbating an inflammatory state and causing further cell death [20,21,22].

Gray matter has been a focus as a potential therapeutic intervention site for ischemic stroke, but the role of white matter damage following ischemia has been neglected. Stroke in areas of the brain containing white matter tracts represents a large minority of patients [23]. Myelin-producing oligodendrocytes create a sheath around axons that allows saltatory conduction. Diffuse excitotoxic death has been observed in oligodendrocytes after ischemia [24,25]. Decreased white matter integrity after ischemic stroke has been associated with long-term cognitive impairment [26]. Additionally, increasing evidence has implicated tissue plasminogen activators’ potentiating effects on ameliorating white matter injury [27]. Edaravone, a potent radical scavenger, was shown to protect oligodendrocyte precursor cells (OPCs) against oxidative stress [28]. Because stem cells possess similar effects on oxidative stress, this may be an avenue by which stem cell transplantation could provide neuroprotection. Adipose-derived mesenchymal cells (ADMSCs) transplanted into the infarct region after ischemia were shown to induce oligodendrocyte progenitor cell (OPC) proliferation in rats [29]. ADMSC administration was also associated with increased myelination [29]. Induced pluripotent stem cell-derived neural stem cell transplantation also increased white matter viability after stroke in pigs [30].

Mechanisms by which stem cells improve recovery after stroke remain unclear, despite many promising studies examining their effects. In the present study, we seek to elucidate the role of the endogenous population of Tregs contained within BMSCs in ischemic stroke and white matter recovery. We hypothesized that depletion of Tregs before BMSC co-culture with OPCs subjected to OGD/R would result in decreased myelination (Figure 1). Furthermore, we expected increased levels of IL-6 in OPCs treated with Treg depleted BMSCs, which would confirm the anti-inflammatory properties of Tregs that have been widely studied. We also predicted that the increase in pro-inflammatory cytokines would affect oligodendrocyte development from OPCs. It is crucial that investigation of the stem cell-brain interaction is pursued, especially the under-explored white matter injury after stroke.

An external file that holds a picture, illustration, etc.
Object name is jcm-08-00537-g001.jpg

T-regulatory cells sequester stroke-induced inflammation leading to less demyelination.

2. Materials and Methods

2.1. Groups

We used 5 groups to assess the effects of BMSCs and Tregs on ischemic OPCs. The first group served as the control and contained OPCs not exposed to oxygen-glucose deprivation (OGD) and not co-cultured with BMSCs (control). The second group contained OPCs exposed to OGD but not co-cultured with BMSCs (OGD). The third group contained OPCs that were exposed to OGD and co-cultured with BMSCs containing Tregs (OPCs with BMSCs Treg+). The fourth group contained OPCs that were exposed to OGD and co-cultured with BMSCs that had been depleted of their Tregs (OPCs with BMSCs Treg). Lastly, the fifth group contained OPCs that were exposed to OGD and co-cultured with BMSCs that were run through the depletion column, but not depleted of their Tregs (OPCs with BMSCs no magnet).

2.2. Cell Culture

Rat OPCs were cultured in vitro as described previously [2]. A total of 40,000 OPCs (ScienCell, Carlsbad, CA, USA) were suspended in 400 μL OPC medium (ScienCell, Carlsbad, CA, USA) and grown in poly-l-lysine-coated glass 8-well plates at 37 °C. The medium contained 500 mL of basal medium, 5 mL of OPC growth supplement and 5 mL of penicillin/streptomycin solution (ScienCell, Carlsbad, CA, USA). Purified BMSCs were obtained and stored in aliquots in dry ice (AllCells, Almeda, CA, USA). After the cells reached confluence, they were subjected to an oxygen-glucose deprivation and reoxygenation (OGD/R) condition (5% CO2, 95% N2) for 90 min. The cells were reoxygenated and co-cultured at 37 °C with BMSCs with or without Tregs, at a concentration of 40,000 cells per well for three more days (Figure 2). OPCs and BMSCs in co-culture were suspended in the same medium and 8-well poly-l-lysine plates were used for the primary culture of OPCs alone. The co-culture was created by directly inoculating the culture medium with the appropriate number of BMSCs with or without their Tregs.

An external file that holds a picture, illustration, etc.
Object name is jcm-08-00537-g002.jpg

Oligodendrocyte progenitor cells (OPCs) were cultured for 3 days and then subjected to oxygen-glucose deprivation and reoxygenation (OGD/R). Bone marrow-derived stem cells (BMSCs) with or without Tregs were then added directly into the cell culture, and both cell types were cultured together for 3 more days.

2.3. Treg Depletion

BMSCs were thawed and run through the depletion column approximately one hour before the co-culture was created. Anti-CD4 and CD25 antibodies were used to label Tregs. The bound antibodies were then conjugated with magnetic microbeads (Miltenyi Biotec, Bergisch Gladbach, Germany). Magnetically labeled cells were isolated by passing the cell suspension through a column containing magnetic metal substrate. CD25+ positive cells were depleted by passing the BMSCs through the column. The resulting eluent was a population of BMSCs that did not contain native Tregs. To mimic the conditions of the OGD with BMSCs Treg group, the OGD with BMSCs no magnet group was similarly run through the depletion column, but the cells were not magnetically labeled and the magnet was inert. We compared this group to the other BMSC groups in order to determine whether the mechanical stress from the depletion column affected the secretion of cytokines or the expression of myelin basic protein (MBP) or O4.

2.4. Measurement of Extracellular Cytokines: IL-6, FGF-β

After OGD/R, culture medium was centrifuged at 3000× g for 15 min and supernatant was collected and frozen at −80 °C. The supernatant was later processed and analyzed using ELISA kits specific to IL-6 (ThermoFisher Scientific, Waltham, MA, USA) and FGF-β (Abcam, Cambridge, MA, USA) with absorbance measured at 450 nm on a Synergy HT plate reader (Bio-Tex Inc., Houston, TX, USA).

2.5. Immunocytochemistry

After cell culture was completed, cells were fixed for 30 min in 4% paraformaldehyde, permeabilized with a 0.1% tween solution, and incubated with fluorescent antibodies with affinity for MBP and oligodendrocyte marker O4 at the manufacturer’s recommended dilutions. Comparison of cell viability between the control and OGD groups was performed using a DAPI nuclear stain (Vector Laboratories, Burlingame, CA, USA). Images of approximately 50 cells/picture were captured in 32 randomly selected areas (1 mm2, n = 32/group) per treatment condition to quantify cell staining for MBP and/or O4 (ImageJ, National Institutes of Health, Bethesda, MD, USA). Stained cells were digitally captured under microscope (20×). In our 8 well plates, one photo was captured from each quadrant in each well (four photos per well). The cells with positively stained nuclei only for MBP and O4 were not counted as MBP+ and O4+ respectively when calculating the percentage of positive cells, as this does not indicate typical morphology. O4+ and MBP+ cells were counted as positive if cells showed processes extending beyond the cell nucleus, as this is consistent with the shape generally assumed by OPCs and oligodendrocytes. Clusters of cells were not considered O4+ or MBP+ if obvious processes extending beyond their nuclei could not be identified.

2.6. Statistics

For categorical independent variables with more than one dependent continuous variable, a one-way ANOVA test was used to determine significance. A p-value less than 0.05 was considered significant. All statistical tests were conducted using IBM SPSS Statistics software (IBM, Armonk, NY, USA). The sample size (eight replicates) was based on our previous study allowing rigorous testing of cultured cells for recognizing a 25% statistically significant difference between treatments and controls [31]. Pairwise comparisons were made with the Tukey-HSD post-hoc test and Fisher’s PLSD post-hoc test for the cell count data and ELISA data respectively.

3. Results

3.1. OGD Resulted in Decreased Cell Viability

Cells were counted using ImageJ software after they were imaged with a confocal microscope. Exposure to OGD/R caused a reduction in number of surviving OPCs to 56.03% of the control (p < 0.05). This survival rate was similar to what was obtained in our previous study [14]. The OPCs co-cultured with the BMSC population that was run through the column but not exposed to the magnet did not show a significantly different cell count than the other BMSC groups, confirming that running the cells through the column did not alter cell survival.

3.2. OPCs Show a More Mature Oligodendrocyte Phenotype

Cells exposed to OGD/R and co-cultured with BMSCs showed greater expression of the O4 marker than both the control and the OGD/R group, suggesting greater maturity (p < 0.001) (Figure 3). There was no significant difference between BMSC groups. Cells that displayed staining only in the nuclear region were not considered positive for the purposes of the quantitative analysis. Cells that did not have processes extending beyond the nucleus staining for either O4 or MBP were unlikely to be of later oligodendrocyte lineage.

An external file that holds a picture, illustration, etc.
Object name is jcm-08-00537-g003.jpg

OPCs co-cultured with BMSCs showed increased expression of intermediate oligodendrocyte marker O4 compared to control. (a) Fluorescently labeled cell nuclei with DAPI, oligodendrocyte progenitor cell (OPC) myelin processes labeled with myelin basic protein, OPC O4 expression labeled with oligo O4, and merge. (b) Quantification of MBP+ cell count in each group shown as percent of cells expressing O4 per image, mean ± SEM.

3.3. Tregs Confer Increased Myelination after OGD/R

Groups co-cultured with BMSCs showed increased myelination compared to groups not co-cultured with BMSCs (p < 0.001), again implying that BMSCs secrete growth factors necessary for oligodendrocyte differentiation (Figure 4). The BMSC Treg+ and BMSC no magnet groups showed increased MBP positive cells compared to the BMSC Treg group (both ps < 0.01). Since the BMSC no magnet group was not depleted of its Tregs, this suggests that the presence of the endogenous Treg population within BMSCs influences the outgrowth and production of myelin processes. Qualitatively, the BMSC Treg+ and BMSC no magnet group had more delineated and robust myelin processes than the BMSC Treg group. No significant differences in staining for either O4 or MBP were seen between the OGD with BMSCs Treg+ group and the OGD with BMSCs no magnet group, and this therefore suggests that running the cells through the column did not alter O4 or MBP expression.

An external file that holds a picture, illustration, etc.
Object name is jcm-08-00537-g004.jpg

BMSCs with Tregs confer increased myelination compared to BMSCs without Tregs. (a) Fluorescently labeled cell nuclei with DAPI, OPC myelin processes labeled with myelin basic protein (MBP), and merge. (b) Quantification of MBP+ cell count in each group shown as percent of cells expressing MBP per image, mean ± SEM.

3.4. Treg+ Groups Showed Increased Extracellular Cytokines

Extracellular secretion of cytokines FGF-β and IL-6 was different among the BMSC groups (p = 0.05 and p < 0.0001, respectively). Interestingly, the BMSC Treg+ showed increased IL-6 production compared to both other groups (both ps < 0.001) (Figure 5). There was increased FGF-β secretion in the BMSC no magnet group compared to the BMSC Treg group (p < 0.05). The BMSC Treg+ group displayed more secretion of FGF-β than the BMSC Treg group, suggesting that the BMSCs were more active in this group (p < 0.05). There was no significant difference between the FGF-β secretion in the BMSC Treg+ group and the BMSC no magnet group (p = 0.72). We assayed both the control and OGD supernatants in both ELISAs, but no results were obtainable because both ELISA kits were customized to detect human cytokines only.

An external file that holds a picture, illustration, etc.
Object name is jcm-08-00537-g005.jpg

BMSCs containing Tregs displayed increased IL-6 secretion. (a) Concentration of interleukin 6 (IL-6) in cell supernatant shown as mean ± SD. (b) Concentration of fibroblast growth factor beta (FGF-β) in cell supernatant shown as mean ± SD.

4. Discussion

The present study tested the effects of Treg cells within a BMSC population on stroke-induced white matter injury in OPCs. Our main findings are as follows: (1) OGD reduced cell viability of OPCs; (2) OPCs treated with BMSCs containing their endogenous Treg population displayed increased myelin production compared to BMSCs with depleted Tregs; (3) co-culture with BMSCs with and without Tregs induced differentiation of immature OPCs into myelin-producing oligodendrocytes compared to control; and (4) extracellular cytokines were increased in the Treg+ groups. In addition, treatment with BMSCs in general (with or without Tregs) was associated with increased OPC differentiation into myelinating oligodendrocytes. This implies that stem cell transplantation into injured white matter may aid in healing and quicker remyelination of damaged tissue after ischemic stroke.

Our results are supported by a plethora of studies showing the beneficial effects of BMSC transplantation on white matter after ischemic stroke. Mice and rats subjected to middle cerebral artery occlusion (MCAO) displayed an increased expression of MBP or O4 following BMSC transplantation compared to the MCAO group devoid of BMSC treatment [32,33]. Similarly, exposure to the secretome of endothelial progenitor cells enhanced OPC differentiation, myelin production and behavioral outcomes in mice subjected to prolonged hypoperfusion [34]. These studies support our finding that OPCs produced more myelin when treated with BMSCs, which may be due to the variety of growth factors secreted by these stem cells cultured in an environment containing ischemic cells [35]. In particular, brain-derived neurotrophic factor (BDNF) has been extensively identified as a major modulator of oligodendrocyte lineage function [36,37,38]. Indeed, BDNF positive Tregs are upregulated compared to other BDNF positive CD4+ T-cells in the human brain following ischemic stroke, suggesting that Tregs may influence oligodendrocyte differentiation via the BDNF pathway [39]. Moreover, BDNF/TrkB activity is postulated to activate sonic hedgehog (Shh) signaling, which mediates oligodendrocyte differentiation [40,41]. A-kinase anchor protein has also been implicated recently as an important modulator of OPC differentiation, implicating that certain proteins may interact with Tregs in directing OPC-mediated myelination [42]. While the results of our study support previous research about white matter injury and the therapeutic benefits of stem cells, they also provide evidence that Tregs act as a key mediator of myelination, perhaps through the BDNF pathway.

Although our previous study showed that Tregs are powerful negative regulators of pro-inflammatory cytokine secretion, specifically IL-6, the results of the current study reveal a unique neuroprotective property of IL-6 in OPCs [14]. We saw an increase in IL-6 levels in the BMSC co-culture group containing Tregs. This may be due to increased BMSC activity in this group, which is supported by our results that there was increased FGF-β secretion in both the BMSC Treg+ and BMSC no magnet group. Although many studies have labeled IL-6 as a pro-inflammatory cytokine, there is equally compelling evidence suggesting that it also has neuroprotective properties and that it is involved in crosstalk with the BDNF pathway [43,44]. Indeed, the treatment of ischemic pheochromocytoma cells with mesenchymal stem cells produces therapeutic effects due to IL-6 and vascular endothelial growth factor secretion [45]. IL-6 has also been shown to confer protection against excitotoxic cell death in neurons [46]. Therefore, IL-6 seems to have dual functions in the brain and more research is warranted to elucidate the effects of this cytokine on ischemic OPCs specifically.

As stated above, the group treated with BMSCs containing Tregs displayed increased MBP production compared to the Treg depleted BMSCs. To date, limited studies demonstrate a potential interaction between Tregs and BDNF/Shh signaling in the brain. Of note, Shh signaling was necessary for expansion of Tregs by dendritic cells, but this was not in the context of the CNS [47]. This information would aid in elucidating the mechanism by which Tregs result in increased MBP expression. Additionally, treatment of cells belonging to the oligodendrocyte lineage showed an increase in myelination after exposure to IL-6 fused with its receptor in a dose-dependent manner [48]. This further supports the notion that IL-6 may have beneficial effects in the CNS and could serve as a positive regulator of OPC differentiation. Moreover, bone marrow-derived-mast cell production of IL-6 was enhanced after exposure to cell-cell contact with Tregs [49]. More specifically, Tregs use the cytokine transforming growth factor beta to modulate levels of IL-6 secreted by mast cells, controlling the inflammatory milieu and increasing neutrophil clearance with increasing levels of IL-6 secretion [50]. Therefore, IL-6 may have anti-inflammatory properties which influence differentiation of OPCs, and Tregs may be direct modulators of its secretion. Effective treatments targeting white matter injury will ensure survival, differentiation and protection of OPCs from inflammation, as this is critical for the remyelination process [51]. More specifically, the complex interactions between BDNF, IL-6, Tregs and BMSCs should be investigated to further elucidate the unique roles that each plays in influencing the oligodendrocyte lineage.

Recognizing that BMSCs are unlikely to transdifferentiate into myelin-producing oligodendrocytes before six days after induction, we did not include stem cell markers in our immunocytochemistry assays in order to distinguish between the original OPCs and stem cells [52]. Future studies investigating the communication between OPCs and BMSCs should include this to better differentiate between the cells and obtain a more definitive cell count. Staining for an earlier OPC lineage marker, such as NG2, in combination with intermediate and later markers would similarly allow characterization of OPCs at all levels of maturity. In this same vein, we were unable to explore the differences in surviving cell number between the groups treated with BMSCs due to their direct introduction into the cell culture. Cell therapy with Tregs or stem cells has shown to result in increased cell viability in neurons and oligodendrocytes after ischemic conditions [14,29,30]. We found that BMSC treatment resulted in healthier oligodendrocytes, but the effects on OPC cell number may be addressed in future experiments. In addition, this study was conducted completely in vitro, and therefore has constraints regarding clinical application. Future studies should focus on examining the in vivo effects of Tregs on white matter injury due to ischemic stroke to segue into the clinic.

In conclusion, we found that BMSC co-culture was associated with increased differentiation of OPCs after ischemic injury and that Treg exposure resulted in more robust expression of MBP. These results confirmed prior research highlighting the beneficial effects of BMSCs after ischemic stroke. Our novel finding consisted of the increased expression of MBP due to exposure to Tregs, but the exact mechanism by which this occurs is unclear at this stage. Limitations including the restricted clinical application and the lack of visualization of stem cell markers should be taken into consideration in future research. However, the finding that Tregs resulted in overall healthier oligodendrocytes emphasizes their potential therapeutic benefit to white matter recovery after ischemic stroke.

Acknowledgments

The authors express their gratitude to all staff of Borlongan’s Neural Transplantation Laboratory who provided critical discussion of the results and extended excellent technical assistance, especially Hung Nguyen.

Author Contributions

Conceptualization, S.Z., E.G.N., and C.V.B.; methodology, Y.K. and C.V.B.; formal analysis, S.Z., Y.K., and C.V.B.; investigation, S.Z., E.G.N., and Y.K.; resources, C.V.B.; data curation, S.Z., E.G.N., and C.V.B..; writing and original draft preparation, S.Z. and C.V.B.; writing, review and editing, S.Z. and C.V.B.; visualization, S.Z., E.G.N., and C.V.B.; supervision, Y.K. and C.V.B.; project administration, C.V.B.; funding acquisition, C.V.B.

Funding

This research was funded by the National Institutes of Health (NIH): NIH R01 NS071956, NIH R01 NS090962, NIH R21 NS089851, NIH R21 NS094087; and by the U.S. Department of Veterans Affairs (VA): VA Merit Review I01 BX001407. Sydney Zarriello received additional funding via the 2018 Summer Scholarly Award Experience from the Office of Research, Innovation & Scholarly Endeavors at the University of South Florida Morsani College of Medicine.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

1. Benjamin E.J., Blaha M.J., Chiuve S.E., Cushman M., Das S.R., Deo R., de Ferranti S.D., Floyd J., Fornage M., Gillespie C., et al. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation. 2017;135:e146–e603. [PMC free article] [PubMed] [Google Scholar]
2. Mehta S.L., Manhas N., Raghubir R. Molecular targets in cerebral ischemia for developing novel therapeutics. Brain Res. Rev. 2007;54:34–66. doi: 10.1016/j.brainresrev.2006.11.003. [PubMed] [CrossRef] [Google Scholar]
3. Liesz A., Dalpke A., Mracsko E., Antoine D.J., Roth S., Zhou W., Yang H., Na S.Y., Akhisaroglu M., Fleming T., et al. DAMP signaling is a key pathway inducing immune modulation after brain injury. J. Neurosci. 2015;35:583–598. doi: 10.1523/JNEUROSCI.2439-14.2015. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
4. Chamorro A., Meisel A., Planas A.M., Urra X., van de Beek D., Veltkamp R. The immunology of acute stroke. Nat. Rev. Neurol. 2012;8:401–410. doi: 10.1038/nrneurol.2012.98. [PubMed] [CrossRef] [Google Scholar]
5. Jin R., Yang G., Li G. Inflammatory mechanisms in ischemic stroke: Role of inflammatory cells. J. Leukoc. Biol. 2010;87:779–789. doi: 10.1189/jlb.1109766. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
6. Borlongan C.V., Rodrigues A.A., Jr., Oliveira M.C. Breaking the barrier in stroke: What should we know? A mini-review. Curr. Pharm. Des. 2012;18:3615–3623. doi: 10.2174/138161212802002670. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
7. Napoli E., Borlongan C.V. Recent Advances in Stem Cell-Based Therapeutics for Stroke. Transl. Stroke Res. 2016;7:452–457. doi: 10.1007/s12975-016-0490-6. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
8. Napoli E., Borlongan C.V. Stem Cell Recipes of Bone Marrow and Fish: Just What the Stroke Doctors Ordered. Stem Cell Rev. 2017;13:192–197. doi: 10.1007/s12015-016-9716-y. [PubMed] [CrossRef] [Google Scholar]
9. Borlongan C.V., Glover L.E., Tajiri N., Kaneko Y., Freeman T.B. The great migration of bone marrow-derived stem cells toward the ischemic brain: Therapeutic implications for stroke and other neurological disorders. Prog. Neurobiol. 2011;95:213–228. doi: 10.1016/j.pneurobio.2011.08.005. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
10. Kfoury Y., Scadden D.T. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell. 2015;16:239–253. doi: 10.1016/j.stem.2015.02.019. [PubMed] [CrossRef] [Google Scholar]
11. Prockop D.J. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276:71–74. doi: 10.1126/science.276.5309.71. [PubMed] [CrossRef] [Google Scholar]
12. Stonesifer C., Corey S., Ghanekar S., Diamandis Z., Acosta S.A., Borlongan C.V. Stem Cell Therapy for Abrogating Stroke-Induced Neuroinflammation and Relevant Secondary Cell Death Mechanisms. Prog. Neurobiol. 2017;158:94–131. doi: 10.1016/j.pneurobio.2017.07.004. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
13. Glover L.E., Tajiri N., Lau T., Kaneko Y., van Loveren H., Borlongan C.V. Immediate, but not delayed, microsurgical skull reconstruction exacerbates brain damage in experimental traumatic brain injury model. PLoS ONE. 2012;7:e33646. doi: 10.1371/journal.pone.0033646. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
14. Neal E.G., Acosta S.A., Kaneko Y., Ji X., Borlongan C.V. Regulatory T-cells within bone marrow-derived stem cells actively confer immunomodulatory and neuroprotective effects against stroke. J. Cereb. Blood Flow Metab. 2018 doi: 10.1177/0271678X18766172. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
15. Williams L.M., Rudensky A.Y. Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires continued expression of Foxp3. Nat. Immunol. 2007;8:277–284. doi: 10.1038/ni1437. [PubMed] [CrossRef] [Google Scholar]
16. Wang G., Shi Y., Jiang X., Leak R.K., Hu X., Wu Y., Pu H., Li W.W., Tang B., Wang Y., et al. HDAC inhibition prevents white matter injury by modulating microglia/macrophage polarization through the GSK3beta/PTEN/Akt axis. Proc. Natl. Acad. Sci. USA. 2015;112:2853–2858. doi: 10.1073/pnas.1501441112. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
17. Suenaga J., Hu X., Pu H., Shi Y., Hassan S.H., Xu M., Leak R.K., Stetler R.A., Gao Y., Chen J. White matter injury and microglia/macrophage polarization are strongly linked with age-related long-term deficits in neurological function after stroke. Exp. Neurol. 2015;272:109–119. doi: 10.1016/j.expneurol.2015.03.021. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
18. Weirather J., Hofmann U.D., Beyersdorf N., Ramos G.C., Vogel B., Frey A., Ertl G., Kerkau T., Frantz S. Foxp3+ CD4+ T cells improve healing after myocardial infarction by modulating monocyte/macrophage differentiation. Circ. Res. 2014;115:55–67. doi: 10.1161/CIRCRESAHA.115.303895. [PubMed] [CrossRef] [Google Scholar]
19. Zhou K., Zhong Q., Wang Y.C., Xiong X.Y., Meng Z.Y., Zhao T., Zhu W.Y., Liao M.F., Wu L.R., Yang Y.R., et al. Regulatory T cells ameliorate intracerebral hemorrhage-induced inflammatory injury by modulating microglia/macrophage polarization through the IL-10/GSK3beta/PTEN axis. J. Cereb. Blood Flow Metab. 2017;37:967–979. doi: 10.1177/0271678X16648712. [PMC free article] [PubMed] [CrossRef] [Google Scholar] Retracted
20. Dolati S., Ahmadi M., Khalili M., Taheraghdam A.A., Siahmansouri H., Babaloo Z., Aghebati-Maleki L., Jadidi-Niaragh F., Younesi V., Yousefi M. Peripheral Th17/Treg imbalance in elderly patients with ischemic stroke. Neurol. Sci. 2018;39:647–654. doi: 10.1007/s10072-018-3250-4. [PubMed] [CrossRef] [Google Scholar]
21. Hu Y., Zheng Y., Wu Y., Ni B., Shi S. Imbalance between IL-17A-producing cells and regulatory T cells during ischemic stroke. Mediators Inflamm. 2014;2014:813045. doi: 10.1155/2014/813045. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
22. Wang X., Xuan W., Zhu Z.Y., Li Y., Zhu H., Zhu L., Fu D.Y., Yang L.Q., Li P.Y., Yu W.F. The evolving role of neuro-immune interaction in brain repair after cerebral ischemic stroke. CNS Neurosci. Ther. 2018;24:1100–1114. doi: 10.1111/cns.13077. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
23. Matute C., Domercq M., Perez-Samartin A., Ransom B.R. Protecting white matter from stroke injury. Stroke. 2013;44:1204–1211. doi: 10.1161/STROKEAHA.112.658328. [PubMed] [CrossRef] [Google Scholar]
24. Tekkok S.B., Goldberg M.P. Ampa/kainate receptor activation mediates hypoxic oligodendrocyte death and axonal injury in cerebral white matter. J. Neurosci. 2001;21:4237–4248. doi: 10.1523/JNEUROSCI.21-12-04237.2001. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
25. Yoshioka A., Yamaya Y., Saiki S., Kanemoto M., Hirose G., Beesley J., Pleasure D. Non-N-methyl-D-aspartate glutamate receptors mediate oxygen–glucose deprivation-induced oligodendroglial injury. Brain Res. 2000;854:207–215. doi: 10.1016/S0006-8993(99)02359-8. [PubMed] [CrossRef] [Google Scholar]
26. Schaapsmeerders P., Tuladhar A.M., Arntz R.M., Franssen S., Maaijwee N.A., Rutten-Jacobs L.C., Schoonderwaldt H.C., Dorresteijn L.D., van Dijk E.J., Kessels R.P., et al. Remote Lower White Matter Integrity Increases the Risk of Long-Term Cognitive Impairment After Ischemic Stroke in Young Adults. Stroke. 2016;47:2517–2525. doi: 10.1161/STROKEAHA.116.014356. [PubMed] [CrossRef] [Google Scholar]
27. Correa F., Gauberti M., Parcq J., Macrez R., Hommet Y., Obiang P., Hernangomez M., Montagne A., Liot G., Guaza C., et al. Tissue plasminogen activator prevents white matter damage following stroke. J. Exp. Med. 2011;208:1229–1242. doi: 10.1084/jem.20101880. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
28. Takase H., Liang A.C., Miyamoto N., Hamanaka G., Ohtomo R., Maki T., Pham L.D., Lok J., Lo E.H., Arai K. Protective effects of a radical scavenger edaravone on oligodendrocyte precursor cells against oxidative stress. Neurosci. Lett. 2018;668:120–125. doi: 10.1016/j.neulet.2018.01.018. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
29. Otero-Ortega L., Gutierrez-Fernandez M., Ramos-Cejudo J., Rodriguez-Frutos B., Fuentes B., Sobrino T., Hernanz T.N., Campos F., Lopez J.A., Cerdan S., et al. White matter injury restoration after stem cell administration in subcortical ischemic stroke. Stem Cell Res. Ther. 2015;6:121. doi: 10.1186/s13287-015-0111-4. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
30. Baker E.W., Platt S.R., Lau V.W., Grace H.E., Holmes S.P., Wang L., Duberstein K.J., Howerth E.W., Kinder H.A., Stice S.L., et al. Induced Pluripotent Stem Cell-Derived Neural Stem Cell Therapy Enhances Recovery in an Ischemic Stroke Pig Model. Sci. Rep. 2017;7:10075. doi: 10.1038/s41598-017-10406-x. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
31. Kaneko Y., Pappas C., Tajiri N., Borlongan C.V. Oxytocin modulates GABAAR subunits to confer neuroprotection in stroke in vitro. Sci. Rep. 2016;6:35659. doi: 10.1038/srep35659. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
32. Yu X., Wu H., Zhao Y., Guo Y., Chen Y., Dong P., Mu Q., Wang X., Wang X. Bone marrow mesenchymal stromal cells alleviate brain white matter injury via the enhanced proliferation of oligodendrocyte progenitor cells in focal cerebral ischemic rats. Brain Res. 2018;1680:127–136. doi: 10.1016/j.brainres.2017.12.019. [PubMed] [CrossRef] [Google Scholar]
33. Zhang J., Li Y., Zhang Z.G., Lu M., Borneman J., Buller B., Savant-Bhonsale S., Elias S.B., Chopp M. Bone marrow stromal cells increase oligodendrogenesis after stroke. J. Cereb. Blood Flow Metab. 2009;29:1166–1174. doi: 10.1038/jcbfm.2009.41. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
34. Maki T., Morancho A., Martinez-San Segundo P., Hayakawa K., Takase H., Liang A.C., Gabriel-Salazar M., Medina-Gutierrez E., Washida K., Montaner J., et al. Endothelial Progenitor Cell Secretome and Oligovascular Repair in a Mouse Model of Prolonged Cerebral Hypoperfusion. Stroke. 2018;49:1003–1010. doi: 10.1161/STROKEAHA.117.019346. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
35. Chen X., Li Y., Wang L., Katakowski M., Zhang L., Chen J., Xu Y., Gautam S.C., Chopp M. Ischemic rat brain extracts induce human marrow stromal cell growth factor production. Neuropathology. 2002;22:275–279. doi: 10.1046/j.1440-1789.2002.00450.x. [PubMed] [CrossRef] [Google Scholar]
36. Miyamoto N., Maki T., Shindo A., Liang A.C., Maeda M., Egawa N., Itoh K., Lo E.K., Lok J., Ihara M., et al. Astrocytes Promote Oligodendrogenesis after White Matter Damage via Brain-Derived Neurotrophic Factor. J. Neurosci. 2015;35:14002–14008. doi: 10.1523/JNEUROSCI.1592-15.2015. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
37. Tsiperson V., Huang Y., Bagayogo I., Song Y., VonDran M.W., DiCicco-Bloom E., Dreyfus C.F. Brain-derived neurotrophic factor deficiency restricts proliferation of oligodendrocyte progenitors following cuprizone-induced demyelination. ASN Neuro. 2015;7 doi: 10.1177/1759091414566878. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
38. Lundegardh G., Hansson L.E., Nyren O., Adami H.O., Krusemo U.B. The risk of gastrointestinal and other primary malignant diseases following gastric cancer. Acta Oncol. 1991;30:1–6. doi: 10.3109/02841869109091804. [PubMed] [CrossRef] [Google Scholar]
39. Chan A., Yan J., Csurhes P., Greer J., McCombe P. Circulating brain derived neurotrophic factor (BDNF) and frequency of BDNF positive T cells in peripheral blood in human ischemic stroke: Effect on outcome. J. Neuroimmunol. 2015;286:42–47. doi: 10.1016/j.jneuroim.2015.06.013. [PubMed] [CrossRef] [Google Scholar]
40. Falcon-Urrutia P., Carrasco C.M., Lois P., Palma V., Roth A.D. Shh Signaling through the Primary Cilium Modulates Rat Oligodendrocyte Differentiation. PLoS ONE. 2015;10:e0133567. doi: 10.1371/journal.pone.0133567. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
41. Ferent J., Zimmer C., Durbec P., Ruat M., Traiffort E. Sonic Hedgehog signaling is a positive oligodendrocyte regulator during demyelination. J. Neurosci. 2013;33:1759–1772. doi: 10.1523/JNEUROSCI.3334-12.2013. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
42. Maki T., Choi Y.K., Miyamoto N., Shindo A., Liang A.C., Ahn B.J., Mandeville E.T., Kaji S., Itoh K., Seo J.H., et al. A-Kinase Anchor Protein 12 Is Required for Oligodendrocyte Differentiation in Adult White Matter. Stem Cells. 2018;36:751–760. doi: 10.1002/stem.2771. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
43. Perigolo-Vicente R., Ritt K., Goncalves-de-Albuquerque C.F., Castro-Faria-Neto H.C., Paes-de-Carvalho R., Giestal-de-Araujo E. IL-6, A1 and A2aR: A crosstalk that modulates BDNF and induces neuroprotection. Biochem. Biophys. Res. Commun. 2014;449:477–482. doi: 10.1016/j.bbrc.2014.05.036. [PubMed] [CrossRef] [Google Scholar]
44. Gu Y., He M., Zhou X., Liu J., Hou N., Bin T., Zhang Y., Li T., Chen J. Endogenous IL-6 of mesenchymal stem cell improves behavioral outcome of hypoxic-ischemic brain damage neonatal rats by supressing apoptosis in astrocyte. Sci. Rep. 2016;6:18587. doi: 10.1038/srep18587. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
45. Lahiani A., Zahavi E., Netzer N., Ofir R., Pinzur L., Raveh S., Arien-Zakay H., Yavin E., Lazarovici P. Human placental eXpanded (PLX) mesenchymal-like adherent stromal cells confer neuroprotection to nerve growth factor (NGF)-differentiated PC12 cells exposed to ischemia by secretion of IL-6 and VEGF. Biochim. Biophys. Acta. 2015;1853:422–430. doi: 10.1016/j.bbamcr.2014.11.009. [PubMed] [CrossRef] [Google Scholar]
46. Ma S.H., Zhuang Q.X., Shen W.X., Peng Y.P., Qiu Y.H. Interleukin-6 reduces NMDAR-mediated cytosolic Ca(2)(+) overload and neuronal death via JAK/CaN signaling. Cell Calcium. 2015;58:286–295. doi: 10.1016/j.ceca.2015.06.006. [PubMed] [CrossRef] [Google Scholar]
47. Holla S., Stephen-Victor E., Prakhar P., Sharma M., Saha C., Udupa V., Kaveri S.V., Bayry J., Balaji K.N. Mycobacteria-responsive sonic hedgehog signaling mediates programmed death-ligand 1- and prostaglandin E2-induced regulatory T cell expansion. Sci. Rep. 2016;6:24193. doi: 10.1038/srep24193. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
48. Valerio A., Ferrario M., Dreano M., Garotta G., Spano P., Pizzi M. Soluble interleukin-6 (IL-6) receptor/IL-6 fusion protein enhances in vitro differentiation of purified rat oligodendroglial lineage cells. Mol. Cell Neurosci. 2002;21:602–615. doi: 10.1006/mcne.2002.1208. [PubMed] [CrossRef] [Google Scholar]
49. Ganeshan K., Bryce P.J. Regulatory T cells enhance mast cell production of IL-6 via surface-bound TGF-beta. J. Immunol. 2012;188:594–603. doi: 10.4049/jimmunol.1102389. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
50. Ganeshan K., Johnston L.K., Bryce P.J. TGF-beta1 limits the onset of innate lung inflammation by promoting mast cell-derived IL-6. J. Immunol. 2013;190:5731–5738. doi: 10.4049/jimmunol.1203362. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
51. Shi H., Hu X., Leak R.K., Shi Y., An C., Suenaga J., Chen J., Gao Y. Demyelination as a rational therapeutic target for ischemic or traumatic brain injury. Exp. Neurol. 2015;272:17–25. doi: 10.1016/j.expneurol.2015.03.017. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
52. Abbaszadeh H.A., Tiraihi T., Delshad A.R., Saghedi Zadeh M., Taheri T. Bone marrow stromal cell transdifferentiation into oligodendrocyte-like cells using triiodothyronine as a inducer with expression of platelet-derived growth factor alpha as a maturity marker. Iran. Biomed. J. 2013;17:62–70. [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Clinical Medicine are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)

-