Bone marrow mesenchymal stromal cells drive protective M2 microglia polarization after brain trauma

Elisa R Zanier, Francesca Pischiutta, Loredana Riganti, Federica Marchesi, Elena Turola, Stefano Fumagalli, Carlo Perego, Emanuela Parotto, Paola Vinci, Pietro Veglianese, Giovanna D'Amico, Claudia Verderio, Maria-Grazia De Simoni, Elisa R Zanier, Francesca Pischiutta, Loredana Riganti, Federica Marchesi, Elena Turola, Stefano Fumagalli, Carlo Perego, Emanuela Parotto, Paola Vinci, Pietro Veglianese, Giovanna D'Amico, Claudia Verderio, Maria-Grazia De Simoni

Abstract

Microglia/macrophages (M) are major contributors to postinjury inflammation, but they may also promote brain repair in response to specific environmental signals that drive classic (M1) or alternative (M2) polarization. We investigated the activation and functional changes of M in mice with traumatic brain injuries and receiving intracerebroventricular human bone marrow mesenchymal stromal cells (MSCs) or saline infusion. MSCs upregulated Ym1 and Arginase-1 mRNA (p < 0.001), two M2 markers of protective M polarization, at 3 and 7 d postinjury, and increased the number of Ym1(+) cells at 7 d postinjury (p < 0.05). MSCs reduced the presence of the lysosomal activity marker CD68 on the membrane surface of CD11b-positive M (p < 0.05), indicating reduced phagocytosis. MSC-mediated induction of the M2 phenotype in M was associated with early and persistent recovery of neurological functions evaluated up to 35 days postinjury (p < 0.01) and reparative changes of the lesioned microenvironment. In vitro, MSCs directly counteracted the proinflammatory response of primary murine microglia stimulated by tumor necrosis factor-α + interleukin 17 or by tumor necrosis factor-α + interferon-γ and induced M2 proregenerative traits, as indicated by the downregulation of inducible nitric oxide synthase and upregulation of Ym1 and CD206 mRNA (p < 0.01). In conclusion, we found evidence that MSCs can drive the M transcriptional environment and induce the acquisition of an early, persistent M2-beneficial phenotype both in vivo and in vitro. Increased Ym1 expression together with reduced in vivo phagocytosis suggests M selection by MSCs towards the M2a subpopulation, which is involved in growth stimulation and tissue repair.

Figures

Fig. 1
Fig. 1
Experimental design of in vivo and in vitro experiments. (A) In vivo experiments: traumatic brain injury (TBI)/sham surgery was done 1 d before treatment. Mesenchymal stromal cells (MSCs) or phosphate buffered saline (PBS) were infused in the contralateral ventricle. Sensorimotor deficits were evaluated at 0, 7, 21, and 35 d. Animals were sacrificed at 3 or 7 d for real time reverse transcription (RT) polymerase chain reaction (PCR) or at 7 or 35 d for histological analysis. (B) In vitro experiments: primary murine microglial cells were cultured for 48 h then, at the timepoints indicated in the plan, were exposed to 1) proinflammatory stimuli [tumor necrosis factor (TNF)-α/interleukin (IL-17) or TNF-α/interferon (IFN)-γ] for M1 classical activation; 2) MSCs; 3) proinflammatory stimuli followed by either direct or indirect (transwell) MSC co-culture. Unexposed cultures served as controls. After 72 h, co-cultures were analyzed by real-time PCR or immunohistochemistry. icv = intracerebroventricular
Fig. 2
Fig. 2
Effects of infusion of mesenchymal stromal cells (MSCs) on sensorimotor deficits. Infusion of MSCs induced early and persistent improvement of sensorimotor deficits, as measured by (A) neuroscore or (B) beam walk tests. (A) Neuroscore test showed sensorimotor improvement in traumatic brain injury (TBI) MSCs mice from 7 d, while (B) the beam walk test showed a significant improvement of TBI MSCs from 21 d. Data are mean ± SD, n = 8, 2-way analysis of variance for RM followed by Tukey’s test. PBS = phosphate buffered saline
Fig. 3
Fig. 3
mRNA expression of genes related to microglia activation and polarization in brain cortices, 3 and 7d after surgery. (AD) CD11b, TNFα, CD86, and CD68 were significantly upregulated in traumatic brain injury (TBI) mice compared with sham-operated mice both at 3 and 7 d after surgery, with no difference between TBI phosphate-buffered saline (PBS) and TBI mesenchymal stromal cells (MSCs) mice. (EH) Ym1, Arginase-1, SOCS3, and CD206 were significantly upregulated in TBI mice compared with sham-operated mice at 3 d, but not at 7 d, after surgery. Infusion of MSCs significantly increased the expression of Ym1 and Arginase-1 in TBI MSCs mice compared with TBI PBS mice at both timepoints considered. No difference in the mRNA expression of SOCS3 was found between TBI PBS mice and TBI MSCs mice. At 7 d, TBI MSCs mice showed increased expression of CD206 compared with sham PBS mice. Data are expressed as the fold induction compared with the sham PBS group. Data are mean ± SD, n = 8. **p < 0.01, ***p < 0.001 versus sham or TBI PBS mice. Two-way analysis of variance followed by Tukey’s test
Fig. 4
Fig. 4
Immunohistochemical analysis of CD45, CD11b, and CD68, and quantification of their co-localization in the injured cortex. Representative micrographs of (A) CD45, (B) CD11b, and (C) CD68 immunostainings and their quantifications 7 d after traumatic brain injury (TBI). The number of CD45high cells was significantly increased in TBI mesenchymal stromal cells (MSCs) mice, whereas no differences were observed in the expression of either CD11b or CD68. Representative micrographs of CD68 (green) and CD11b (red) co-localization in (D) TBI phosphate-buffered saline (PBS) and (D’) TBI MSCs mice. (D, merge) In TBI PBS mice, CD68 often co-localized with the membrane marker CD11b, while in TBI MSCs mice (D’, merge) it remained mainly located in the cytoplasm, thus yielding less co-localization with CD11b. (E, E’) Quantification of co-localized voxels in the 3-dimensional confocal acquisitions showed a reduction of CD68/CD11b-positive voxels after infusion of MSCs, indicating a reduction of lysosomal activity in TBI MSCs mice compared with TBI PBS mice (F). Data are mean ± SD, n = 8 (A, B, E). *p < 0.05, unpaired t test. Bars = 20 μm
Fig. 5
Fig. 5
Immunohistochemical analysis of Ym1 in the injured cortex and quantification of its co-localization with CD68 in the injured hippocampus. Representative micrographs of (A) Ym1 immunostaining 7 d after traumatic brain injury (TBI) in phosphate-buffered saline (PBS)- or mesenchymal stromal cells (MSCs) treated mice, and its related quantification, which shows an increase in the expression of Ym1 in TBI MSCs mice compared with TBI PBS mice. Co-localization of Ym1 (purple) and CD68 (green) in (B) TBI PBS and (C) TBI MSCs mice. Bar = 20 μm. (D, E) Quantification of co-localized voxels in the 3-dimensional (3D) confocal acquisitions showed a reduction of (F) Ym1/CD68-positive voxels after infusion of MSCs. Triple immunofluorescence for CD11b (red), CD68 (green), and Ym1 (purple) for (G) TBI PBS and (H) TBS MSCs mice. (G) In TBI PBS mice, co-localization between Ym1 and CD68 (white) is observed in cells with strong co-localization between CD68 and CD11b (yellow). Xyz-view and 3D renderings of co-localized pixels (centre and right panels) better illustrate this. (H) In TBI MSCs mice, cells with reduced Ym1/CD68 co-localization (white) show diminished CD68/CD11b co-localization. The Xyz-view and 3D renderings of co-localized pixels are shown in centre and right panels in (H). Bar = 5 μm. Data are mean ± SD, n = 8 (A, F). *p < 0.05, unpaired t test
Fig. 6
Fig. 6
Localization of CD11b/Ym1 double-positive cells with PKH26-labeled mesenchymal stromal cells (MSCs) in the injured cortex. (A) At 7 d after traumatic brain injury, in mice infused with MSCs, the cells positive for CD11b (green) and Ym1 (purple) showed direct contact with infused MSCs (PKH26, red, A), as better depicted in (B) the 3-dimensional rendering. Bar = 20 μm
Fig. 7
Fig. 7
mRNA expression of cytokines and growth factors in brain cortices, 3 and 7 d after surgery. (AD) CCL2, IL-1β, IL-10, and IGF1 were significantly upregulated in traumatic brain injury (TBI) compared with sham-operated mice at 3 d after surgery. (ABD) The upregulation of CCL2, IL-1β, and IGF1 in TBI mice persisted at 7 d after surgery. (AD) There was no difference in the expression of CCL2, IL-1β, IL-10, and IGF1 between TBI phosphate buffered saline (PBS) and TBI MSCs mice at 3 d. (AC) At 7 d, mesenchymal stromal cells (MSCs) infusion significantly increased the expression of CCL2, IL-1β, and IL-10 in TBI MSCs mice compared with TBI PBS mice, while (D) no difference was found in the expression of IGF1. (E) The expression of VEGF was downregulated in TBI PBS mice compared with sham-operated groups both at 3 d and 7 d. TBI MSCs mice showed a trend toward an increase in the expression of VEGF at 7d, restoring VEGF expression close to the levels observed in sham-operated animals. (F) The expression of GFAP was significantly upregulated in TBI mice compared with sham-operated mice at both 3 and 7 d after surgery. Infusion of MSCs induced a significant reduction in the expression of GFAP at 7 d after surgery. Data are shown as fold induction compared with sham PBS group and are mean ± SD, n = 8. **p < 0.01, ***p < 0.001 versus sham or TBI PBS, 2-way analysis of variance followed by Tukey’s test
Fig. 8
Fig. 8
Localization of PKH26-labeled mesenchymal stromal cells (MSCs) and immunofluorescence for growth associated protein 43 (GAP-43) and Ym1 in the injured cortex. Representative micrographs at low magnification showing the presence of PKH26-labeled MSCs (red) in cortical areas positive for GAP-43 (green) at (A) 7 d or (B) 35 d after traumatic brain injury (TBI; nuclei are in blue, bar = 100 μm). PKH26 was visible only at 7 d in mice infused with MSCs, while no PKH26 positivity was detectable at 35 d in either phosphate-buffered saline (PBS)- or MSC-treated mice. GAP-43 appeared to be increased at 7 and 35 d in mice receiving MSCs. (C) At 7 d, in TBI MSCs mice, PKH26-positive cells were found either in association with GAP-43-positive cells or in areas negative for GAP-43. (D) When MSCs reached the neurogenic niche in the subventricular zone, they localized close to GAP-43-positive cells. (E) M2 polarized cells (Ym1-positive, purple) were located in areas positive for GAP-43 and showed strong association with GAP-43-positive cells [(F) 3-dimensional rendering]. Bars in (CE) = 20 μm. DAPI = 4’6-diamidino-2-phenylindole; LV = lateral ventricle
Fig. 9
Fig. 9
In vitro analysis of microglia markers after exposure to mesenchymal stromal cells (MSCs). Microglia expression of (A, B) Ym1 and (C, D) CD206 in control conditions or in co-culture with MSCs for 72 h. (A, C) Quantification of mRNA expression indicates an upregulation of both M2 markers induced by MSC co-culture. Representative confocal images of pure microglial cultures and microglia co-cultured with MSCs stained for (B) Ym1 and (D) CD206. Data are mean ± SD from three independent experiments.*p < 0.05, **p < 0.01, unpaired t test. CTRL = control
Fig. 10
Fig. 10
In vitro analysis of M1 and M2 polarization markers following exposure to proinflammatory stimuli and mesenchymal stromal cells (MSCs). mRNA expression for (A) iNOS, (B) Ym1, and (C) CD206 in control or activated [exposed to tumor necrosis factor (TNF)-α/interleukin (IL)-17 or TNF-α/interferon (IFN)-γ] microglia, maintained in vitro in isolation or co-cultured with MSCs. Data are mean ± SD from 3 independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001 2-way analysis of variance followed by Tukey’s test

References

    1. Lingsma HF, Roozenbeek B, Steyerberg EW, Murray GD, Maas AIR. Early prognosis in traumatic brain injury: from prophecies to predictions. Lancet Neurol. 2010;9:543–554. doi: 10.1016/S1474-4422(10)70065-X.
    1. Xiong Y, Mahmood A, Chopp M. Neurorestorative treatments for traumatic brain injury. Discov Med. 2010;10:434–442.
    1. Christie KJ, Turnley AM. Regulation of endogenous neural stem/progenitor cells for neural repair-factors that promote neurogenesis and gliogenesis in the normal and damaged brain. Front Cell Neurosci. 2012;6:70. doi: 10.3389/fnhum.2012.00028.
    1. Xiong Y, Mahmood A, Meng Y, et al. Delayed administration of erythropoietin reducing hippocampal cell loss, enhancing angiogenesis and neurogenesis, and improving functional outcome following traumatic brain injury in rats: comparison of treatment with single and triple dose. J Neurosurg. 2010;113:598–608. doi: 10.3171/2009.9.JNS09844.
    1. Loane DJ, Faden AI. Neuroprotection for traumatic brain injury: translational challenges and emerging therapeutic strategies. Trends Pharmacol Sci. 2010;31:596–604. doi: 10.1016/j.tips.2010.09.005.
    1. Maas AIR, Stocchetti N, Bullock R. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 2008;7:728–741. doi: 10.1016/S1474-4422(08)70164-9.
    1. Ohtaki H, Ylostalo JH, Foraker JE, et al. Stem/progenitor cells from bone marrow decrease neuronal death in global ischemia by modulation of inflammatory/immune responses. Proc Natl Acad Sci U S A. 2008;105:14638–14643. doi: 10.1073/pnas.0803670105.
    1. Sarnowska A, Braun H, Sauerzweig S, Reymann KG. The neuroprotective effect of bone marrow stem cells is not dependent on direct cell contact with hypoxic injured tissue. Exp Neurol. 2009;215:317–327. doi: 10.1016/j.expneurol.2008.10.023.
    1. Zanier ER, Montinaro M, Vigano M, et al. Human umbilical cord blood mesenchymal stem cells protect mice brain after trauma. Crit Care Med. 2011;39:2501–2510. doi: 10.1097/CCM.0b013e31822629ba.
    1. Nakajima H, Uchida K, Guerrero AR, et al. Transplantation of mesenchymal stem cells promotes an alternative pathway of macrophage activation and functional recovery after spinal cord injury. J Neurotrauma. 2012;29:1614–1625. doi: 10.1089/neu.2011.2109.
    1. Kumar A, Loane DJ. Neuroinflammation after traumatic brain injury: Opportunities for therapeutic intervention. Brain Behav Immun. 2012;26:1191–1201. doi: 10.1016/j.bbi.2012.06.008.
    1. Kumar A, Stoica BA, Sabirzhanov B, Burns MP, Faden AI, Loane DJ. Traumatic brain injury in aged animals increases lesion size and chronically alters microglial/macrophage classical and alternative activation states. Neurobiol Aging. 2013;34:1397–1411. doi: 10.1016/j.neurobiolaging.2012.11.013.
    1. Shechter R, Schwartz M. Harnessing monocyte-derived macrophages to control central nervous system pathologies: no longer “if” but “how”. J Pathol. 2013;229:332–346. doi: 10.1002/path.4106.
    1. Lai AY, Todd KG. Differential regulation of trophic and proinflammatory microglial effectors is dependent on severity of neuronal injury. Glia. 2008;56:259–270. doi: 10.1002/glia.20610.
    1. Madinier A, Bertrand N, Mossiat C, et al. Microglial involvement in neuroplastic changes following focal brain ischemia in rats. PLoS ONE. 2009;4:e8101. doi: 10.1371/journal.pone.0008101.
    1. Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci. 2009;29:13435–13444. doi: 10.1523/JNEUROSCI.3257-09.2009.
    1. Longhi L, Perego C, Ortolano F, et al. Tumor necrosis factor in traumatic brain injury: effects of genetic deletion of p55 or p75 receptor. J Cereb Blood Flow Metab. 2013;33:1182–1189. doi: 10.1038/jcbfm.2013.65.
    1. Chhor V, Le Charpentier T, Lebon S, et al. Characterization of phenotype markers and neuronotoxic potential of polarised primary microglia in vitro. Brain Behav Immun. 2013;32:70–85. doi: 10.1016/j.bbi.2013.02.005.
    1. Hu X, Li P, Guo Y, et al. Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke. 2012;43:3063–3070. doi: 10.1161/STROKEAHA.112.659656.
    1. Fumagalli S, Perego C, Ortolano F, De Simoni M-G. CX3CR1 deficiency induces an early protective inflammatory environment in ischemic mice. Glia. 2013;61:827–842. doi: 10.1002/glia.22474.
    1. Pischiutta F, D’Amico G, Dander E, et al. Immunosuppression does not affect human bone marrow mesenchymal stromal cell efficacy after transplantation in traumatized mice brain. Neuropharmacology. 2014;79:119–126. doi: 10.1016/j.neuropharm.2013.11.001.
    1. Dander E, Lucchini G, Vinci P, et al. Mesenchymal stromal cells for the treatment of graft-versus-host disease: understanding the in vivo biological effect through patient immune monitoring. Leukemia. 2012;26:1681–1684. doi: 10.1038/leu.2011.384.
    1. Zanier ER, Pischiutta F, Villa P, et al. Six-month ischemic mice show sensorimotor and cognitive deficits associated with brain atrophy and axonal disorganization. CNS Neurosci Ther. 2013;19:695–704. doi: 10.1111/cns.12128.
    1. Ortolano F, Colombo A, Zanier ER, et al. c-Jun N-terminal kinase pathway activation in human and experimental cerebral contusion. J Neuropathol Exp Neurol. 2009;68:964–971. doi: 10.1097/NEN.0b013e3181b20670.
    1. Paxinos G, Franklin KBJ. The mouse brain in stereotaxic coordinates. Academic Press, San Diego, CA, USA, 2004.
    1. Capone C, Frigerio S, Fumagalli S, et al. Neurosphere-derived cells exert a neuroprotective action by changing the ischemic microenvironment. PLoS ONE. 2007;2:e373. doi: 10.1371/journal.pone.0000373.
    1. Donnelly DJ, Gensel JC, Ankeny DP, van Rooijen N, Popovich PG. An efficient and reproducible method for quantifying macrophages in different experimental models of central nervous system pathology. J Neurosci Methods. 2009;181:36–44. doi: 10.1016/j.jneumeth.2009.04.010.
    1. Schindelin J, Arganda-Carreras I, Frise E, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9:676–682. doi: 10.1038/nmeth.2019.
    1. Gesuete R, Storini C, Fantin A, et al. Recombinant C1 inhibitor in brain ischemic injury. Ann Neurol. 2009;66:332–342. doi: 10.1002/ana.21740.
    1. Curtis R, Hardy R, Reynolds R, Spruce BA, Wilkin GP. Down-regulation of GAP-43 During Oligodendrocyte Development and Lack of Expression by Astrocytes In Vivo: Implications for Macroglial Differentiation. Eur J Neurosci. 1991;3:876–886. doi: 10.1111/j.1460-9568.1991.tb00099.x.
    1. Riglar DT, Rogers KL, Hanssen E, et al. Spatial association with PTEX complexes defines regions for effector export into Plasmodium falciparum-infected erythrocytes. Nat Commun. 2013;4:1415. doi: 10.1038/ncomms2449.
    1. Longhi L, Perego C, Ortolano F, et al. C1-inhibitor attenuates neurobehavioral deficits and reduces contusion volume after controlled cortical impact brain injury in mice. Crit Care Med. 2009;37:659–665. doi: 10.1097/CCM.0b013e318195998a.
    1. Verderio C, Muzio L, Turola E, et al. Myeloid microvesicles are a marker and therapeutic target for neuroinflammation. Ann Neurol. 2012;72:610–624. doi: 10.1002/ana.23627.
    1. Stein VM, Baumgärtner W, Schröder S, Zurbriggen A, Vandevelde M, Tipold A. Differential expression of CD45 on canine microglial cells. J Vet Med A Physiol Pathol Clin Med. 2007;54:314–320. doi: 10.1111/j.1439-0442.2007.00926.x.
    1. 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 Neuroinflammation. 2011;8:174. doi: 10.1186/1742-2094-8-174.
    1. Ramprasad MP, Terpstra V, Kondratenko N, Quehenberger O, Steinberg D. Cell surface expression of mouse macrosialin and human CD68 and their role as macrophage receptors for oxidized low density lipoprotein. Proc Natl Acad Sci U S A. 1996;93:14833–14838. doi: 10.1073/pnas.93.25.14833.
    1. Kurushima H, Ramprasad M, Kondratenko N, Foster DM, Quehenberger O, Steinberg D. Surface expression and rapid internalization of macrosialin (mouse CD68) on elicited mouse peritoneal macrophages. J Leukoc Biol. 2000;67:104–108.
    1. David S, Kroner A. Repertoire of microglial and macrophage responses after spinal cord injury. Nat Rev Neurosci. 2011;12:388–399. doi: 10.1038/nrn3053.
    1. Franquesa M, Hoogduijn MJ, Reinders ME, et al. Mesenchymal Stem Cells in Solid Organ Transplantation (MiSOT) Fourth Meeting: lessons learned from first clinical trials. Transplantation. 2013;96:234–238. doi: 10.1097/TP.0b013e318298f9fa.
    1. Lambertsen KL, Clausen BH, Babcock AA, et al. Microglia protect neurons against ischemia by synthesis of tumor necrosis factor. J Neurosci. 2009;29:1319–1330. doi: 10.1523/JNEUROSCI.5505-08.2009.
    1. Sierra A, Encinas JM, Deudero JJP, et al. Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem Cell. 2010;7:483–495. doi: 10.1016/j.stem.2010.08.014.
    1. Denes A, Vidyasagar R, Feng J, et al. Proliferating resident microglia after focal cerebral ischaemia in mice. J Cereb Blood Flow Metab. 2007;27:1941–1953. doi: 10.1038/sj.jcbfm.9600495.
    1. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122:787–795. doi: 10.1172/JCI59643.
    1. Walker PA, Bedi SS, Shah SK, et al. Intravenous multipotent adult progenitor cell therapy after traumatic brain injury: modulation of the resident microglia population. J Neuroinflammation. 2012;9:228. doi: 10.1186/1742-2094-9-228.
    1. Giunti D, Parodi B, Usai C, et al. Mesenchymal stem cells shape microglia effector functions through the release of CX3CL1. Stem Cells. 2012;30:2044–2053. doi: 10.1002/stem.1174.
    1. Kim Y-J, Park H-J, Lee G, et al. Neuroprotective effects of human mesenchymal stem cells on dopaminergic neurons through anti-inflammatory action. Glia. 2009;57:13–23. doi: 10.1002/glia.20731.
    1. Micklem K, Rigney E, Cordell J, et al. A human macrophage-associated antigen (CD68) detected by six different monoclonal antibodies. Br J Haematol. 1989;73:6–11. doi: 10.1111/j.1365-2141.1989.tb00210.x.
    1. Holness CL, Simmons DL. Molecular cloning of CD68, a human macrophage marker related to lysosomal glycoproteins. Blood. 1993;81:1607–1613.
    1. Travaglione S, Falzano L, Fabbri A, Stringaro A, Fais S, Fiorentini C. Epithelial cells and expression of the phagocytic marker CD68: scavenging of apoptotic bodies following Rho activation. Toxicol In Vitro. 2002;16:405–411. doi: 10.1016/S0887-2333(02)00028-0.
    1. Neher JJ, Neniskyte U, Zhao J-W, Bal-Price A, Tolkovsky AM, Brown GC. Inhibition of microglial phagocytosis is sufficient to prevent inflammatory neuronal death. J Immunol. 2011;186:4973–4983. doi: 10.4049/jimmunol.1003600.
    1. Neher JJ, Neniskyte U, Brown GC. Primary phagocytosis of neurons by inflamed microglia: potential roles in neurodegeneration. Front Pharmacol. 2012;3:27. doi: 10.3389/fphar.2012.00027.
    1. Bonilla C, Zurita M, Otero L, Aguayo C, Vaquero J. Delayed intralesional transplantation of bone marrow stromal cells increases endogenous neurogenesis and promotes functional recovery after severe traumatic brain injury. Brain Inj. 2009;23:760–769. doi: 10.1080/02699050903133970.
    1. Mahmood A, Lu D, Qu C, Goussev A, Chopp M. Treatment of traumatic brain injury with a combination therapy of marrow stromal cells and atorvastatin in rats. Neurosurgery. 2007;60:546–553. doi: 10.1227/01.NEU.0000255346.25959.99.
    1. Qu C, Mahmood A, Lu D, Goussev A, Xiong Y, Chopp M. Treatment of traumatic brain injury in mice with marrow stromal cells. Brain Res. 2008;1208:234–239. doi: 10.1016/j.brainres.2008.02.042.
    1. Sato A, Ohtaki H, Tsumuraya T, et al. Interleukin-1 participates in the classical and alternative activation of microglia/macrophages after spinal cord injury. J Neuroinflammation. 2012;9:65. doi: 10.1186/1742-2094-9-65.
    1. Babcock AA, Kuziel WA, Rivest S, Owens T. Chemokine expression by glial cells directs leukocytes to sites of axonal injury in the CNS. J Neurosci. 2003;23:7922–7930.
    1. Si Y, Tsou C-L, Croft K, Charo IF. CCR2 mediates hematopoietic stem and progenitor cell trafficking to sites of inflammation in mice. J Clin Invest. 2010;120:1192–1203. doi: 10.1172/JCI40310.
    1. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889–896. doi: 10.1038/ni.1937.
    1. Butovsky O, Bukshpan S, Kunis G, Jung S, Schwartz M. Microglia can be induced by IFN-gamma or IL-4 to express neural or dendritic-like markers. Mol Cell Neurosci. 2007;35:490–500. doi: 10.1016/j.mcn.2007.04.009.
    1. Cho HH, Kim YJ, Kim JT, et al. The role of chemokines in proangiogenic action induced by human adipose tissue-derived mesenchymal stem cells in the murine model of hindlimb ischemia. Cell Physiol Biochem. 2009;24:511–518. doi: 10.1159/000257495.

Source: PubMed

3
Tilaa