A cord blood monocyte-derived cell therapy product accelerates brain remyelination

Arjun Saha, Susan Buntz, Paula Scotland, Li Xu, Pamela Noeldner, Sachit Patel, Amy Wollish, Aruni Gunaratne, Tracy Gentry, Jesse Troy, Glenn K Matsushima, Joanne Kurtzberg, Andrew E Balber, Arjun Saha, Susan Buntz, Paula Scotland, Li Xu, Pamela Noeldner, Sachit Patel, Amy Wollish, Aruni Gunaratne, Tracy Gentry, Jesse Troy, Glenn K Matsushima, Joanne Kurtzberg, Andrew E Balber

Abstract

Microglia and monocytes play important roles in regulating brain remyelination. We developed DUOC-01, a cell therapy product intended for treatment of demyelinating diseases, from banked human umbilical cord blood (CB) mononuclear cells. Immunodepletion and selection studies demonstrated that DUOC-01 cells are derived from CB CD14+ monocytes. We compared the ability of freshly isolated CB CD14+ monocytes and DUOC-01 cells to accelerate remyelination of the brains of NOD/SCID/IL2Rγnull mice following cuprizone feeding-mediated demyelination. The corpus callosum of mice intracranially injected with DUOC-01 showed enhanced myelination, a higher proportion of fully myelinated axons, decreased gliosis and cellular infiltration, and more proliferating oligodendrocyte lineage cells than those of mice receiving excipient. Uncultured CB CD14+ monocytes also accelerated remyelination, but to a significantly lesser extent than DUOC-01 cells. Microarray analysis, quantitative PCR studies, Western blotting, and flow cytometry demonstrated that expression of factors that promote remyelination including PDGF-AA, stem cell factor, IGF1, MMP9, MMP12, and triggering receptor expressed on myeloid cells 2 were upregulated in DUOC-01 compared to CB CD14+ monocytes. Collectively, our results show that DUOC-01 accelerates brain remyelination by multiple mechanisms and could be beneficial in treating demyelinating conditions.

Figures

Figure 1. Severe demyelination of the midline…
Figure 1. Severe demyelination of the midline corpus callosum (CC) area and glial infiltration of NSG mouse brain by cuprizone feeding.
(A) LFB-PAS staining of NSG mice brain after 5 weeks of feeding with (right panel) and without (left panel) 0.2% cuprizone (CPZ). The midline CC area is shown by dotted red boxes in the top panels and then shown at higher magnification in the lower panels. Myelinated axons in the CC of mice fed normal laboratory chow are stained blue. Demyelination of the midline CC region of CPZ-treated animals is shown by the absence of the blue-colored fibers. Scale bars: 2,000 μm (×20 magnification) and 100 μm (×400 magnification). (B) Myelin basic protein immunostaining (green) after 5 weeks of feeding without (left panel) and with CPZ (right panel). Two different magnifications (top row is ×100 and bottom row is ×400) of the CC areas are shown. CC areas are shown by white dotted lines. (C) Immunostaining with microglial marker Iba1 (red, upper panels) and astrocyte marker GFAP (pink, lower panels) after 5 weeks of feeding without (left panels) and with CPZ (right panels). CC areas are shown by white dotted lines. Scale bars: 200 μm. (D) Quantitative analysis of area covered by Iba1-positive (upper panel) and GFAP-positive (lower panel) cells, indicative of their numbers, along the CC. Both Iba1-positive and GFAP-positive cell numbers were significantly higher in the CPZ-treated animals. *P < 0.02, **P < 0.004. n = 3 mice per group . C, control. Data are presented as the mean ± SEM.
Figure 2. Some DUOC-01 cells disseminated from…
Figure 2. Some DUOC-01 cells disseminated from the injection site and persisted in the brain for up to 1 week after intracranial injection.
Cuprizone-fed (CPZ-fed) mice were stereotactically injected with CFSE-labeled DUOC-01 cells. All cell nuclei were stained with DAPI (blue). (A) CFSE-labeled (green) DUOC-01 cells were found in numerous parts of the brain including the injection site. Scale bars: 200 μm. CC, corpus callosum; SV, subventricular. (B) Representative images of CFSE-positive (green) and human nuclei (HuN, red)-positive cells in the brain at the injection site 4 days after injection. Upper left panel is CFSE (green) channel only, lower left panel is HuN (red) channel only, right panel is merge of CFSE, HuN, and DAPI channels. (C) Upper left panel is CFSE channel only, lower left panel is HuN channel only, right panel is merge of CFSE, HuN, and DAPI channels showing presence of DUOC-01 cells 7 days after injection at the CC. (D) Upper left panel is CFSE channel only, lower left panel is HuN channel only, right panel is merge of CFSE, HuN, and DAPI channels showing presence of DUOC-01 cells deep (white arrow) into the brain parenchyma. Scale bars (BD): 100 μm.
Figure 3. LFB-PAS staining analysis of effect…
Figure 3. LFB-PAS staining analysis of effect of DUOC-01 treatment on remyelination following cessation of cuprizone (CPZ) treatment.
(A) LFB-PAS staining 1 week after intracranial injection of CD14+ monocytes (lower panels), DUOC-1 cells (middle panels), or Ringer’s solution (upper panels) in CPZ-fed NSG mice. Midline corpus callosum (CC) area is shown by dotted green line. Scale bars: 2,000 μm (×20 magnification) and 100 μm (×400 magnification). (B) Myelination score based on LFB-PAS staining of mice fed normal chow (control) or CPZ for 5 weeks 1 week after treatment of CPZ-treated mice with CD14+ monocytes, DUOC-01 cells, or Ringer’s. DUOC-01 treatment for 1 week significantly increased the myelination in the CC area compared to Ringer’s-injected controls. **P < 5.962 × 10–5 for this study. The CD14+ cell–treated sample showed an increased amount of remyelination compared to the Ringer’s-treated group, but it was significantly less than the DUOC-01–treated group. *P < 0.003875. Data are presented as the mean ± SEM. Statistical comparisons were performed using the Wilcoxon rank-sum test for clustered data using the clusrank package in R.
Figure 4. Immunostaining analysis of the effect…
Figure 4. Immunostaining analysis of the effect of DUOC-01 treatment on remyelination following cessation of cuprizone (CPZ) treatment.
In all images, myelin basic protein (MBP) staining is shown in green. (A) Representative ×400 laser confocal images of sections of the corpus callosum (CC) area of CPZ-fed mice immunostained with antibodies against MBP (green) and neurofilament-H (NFH, purple), 1 week after treatment with Ringer’s solution (A), DUOC-01 (B), or CD14+ (C). Upper left panels show MBP (green channel), lower left panels show NFH (purple channel), and right panels show enlarged merge images of MBP and NFH channels. Scale bars: 100 μm.
Figure 5. Electron microscopic analysis of remyelination…
Figure 5. Electron microscopic analysis of remyelination status upon DUOC-01 treatment.
Representative ×2,650 (upper panels) and ×8,800 (lower panels) electron micrographs of corpus callosum region of cuprizone-fed mice 1 week after injection of Ringer’s solution (left panels) or DUOC-01 cells (right panels). Blue arrows indicate unmyelinated axons. Red arrowheads indicate mitochondria; enlarged mitochondria are clearly visible in the Ringer’s-treated group. Scale bars: 2.0 μm.
Figure 6. Morphometric analysis of electron micrographs…
Figure 6. Morphometric analysis of electron micrographs of corpus callosum regions of DUOC-01– and Ringer’s-treated mice.
(A) Number of myelinated axons present per ×8,800 electron microscopy field. Data are presented as the mean ± SEM showing all the data points. *P ≤ 4.29 × 10–9. (B) Average number of turns of myelin sheath around axons, with right panels showing a representative electron micrograph of the myelin turns in an axon. *P ≤ 3.4 × 10–6. Scale bars: 100 nm. (C) Scatter plot of g-ratios, showing axonal measurements from 3 different animals in each group. Horizontal lines indicate mean g-ratios. P ≤ 0.014. (D) Average size of mitochondria (area in nm2). Mean difference is significant between DUOC-01 and Ringer’s groups. *P ≤ 9.3 × 10–5. (E) Average number of mitochondria per ×8,800 field. The mean difference is significant between DUOC-01 and Ringer’s groups. *P ≤ 0.02. Each column represents the value of measurements from 3 different animals. Error bars indicate the SEM. Statistical comparisons were performed using an unpaired 2-tailed Student’s t test.
Figure 7. DUOC-01 cell treatment reduces severe…
Figure 7. DUOC-01 cell treatment reduces severe astrogliosis and microglial infiltration.
(A) A quantitative cellularity scoring of LFB-stained brain slices on a scale of 0 to 3. **P ≤ 7.618 × 10–5, n ≥ 5. Control, not cuprizone fed; CPZ, cuprizone fed; Ringer’s, 1 week after Ringer’s injection; DUOC-01, 1 week after DUOC-01 injection. Data are presented as the mean ± SEM showing each data point. Statistical comparisons were performed using the Wilcoxon rank-sum test for clustered data using the clusrank package in R. (B) Cellularity status by immunostaining using astrocyte-specific (GFAP, right panels) and microglia-specific (Iba1, left panels) markers. Midline corpus callosum (CC) areas are shown in dotted line. Scale bars: 100 μm. (C) Quantitative analysis of area covered by Iba1-positive (upper panel) and GFAP-positive (lower panel) cells, indicative of their numbers, along the CC. Both the numbers of Iba1-positive (microglia) and GFAP-positive (astrocytes) cells were significantly lower in the DUOC-01–treated mice. *P < 0.002; **P < 0.01. n = 3 mice per group. Areas covered by each channel (either GFAP or Iba1) per microscopic field were quantified by ImageJ software. Data are presented as the mean ± SEM. Statistical comparisons were performed using an unpaired 2-tailed Student’s t test.
Figure 8. DUOC-01 treatment promotes oligodendrocyte proliferation.
Figure 8. DUOC-01 treatment promotes oligodendrocyte proliferation.
(A) Representative image of corpus callosum area of brains of cuprizone-fed mice treated with DUOC-01 cells (lower panels) or Ringer’s solution (upper panels) stained with antibodies against Olig2 and Ki67. Yellow arrows indicate nuclei positive for both Olig2 and Ki67, blue arrows indicate only Ki67-positive nuclei. Scale bars: 50 μm. (B) Average number of Olig2+Ki67+ cells (indicating proliferating oligodendrocytes) present per ×400 microscopic field were significantly higher in DUOC-01–treated samples compared to the Ringer’s control. *P < 0.01. Statistical comparisons were performed using an unpaired 2-tailed Student’s t test.
Figure 9. Comparative whole-transcriptome analysis of CD14…
Figure 9. Comparative whole-transcriptome analysis of CD14 and DUOC-01 cells.
(A) Venn diagram displaying the findings from microarray analysis showing the number of genes differentially expressed in purified fresh CD14+ (n = 4) and DUOC-01 (n = 3) cells as well as genes expressed by both cell types. MAS5-normalized data were used to filter out expressed/nonexpressed genes. The figure represents the most stringent analysis; to be scored as expressed, the transcript had to be detected above background in all samples of a given cell type analyzed. See the text for expression figures at different stringencies. (B) Volcano plot depiction of findings from microarray analysis showing the genes differentially expressed in purified fresh CD14+ and DUOC-01 cells. The log10 of Bonferroni-Hochberg–corrected P values in ANOVA (y axis) is plotted against the of fold change between 2 groups (x axis). Red lines delineate the cutoffs for genes significantly (P < 0.05) downregulated (left) or upregulated (right) in DUOC-01 cells. Each data point represents 1 gene probe set. (C) Heat maps showing differentially expressed genes. Up- and downregulated genes are displayed in red and blue, respectively. There were 9,645 genes that are differentially expressed at a magnitude of at least 2-fold.

References

    1. Nayak D, Roth TL, McGavern DB. Microglia development and function. Annu Rev Immunol. 2014;32:367–402. doi: 10.1146/annurev-immunol-032713-120240.
    1. Ginhoux F, Lim S, Hoeffel G, Low D, Huber T. Origin and differentiation of microglia. Front Cell Neurosci. 2013;7:e86667.
    1. Matsushima GK, et al. Absence of MHC class II molecules reduces CNS demyelination, microglial/macrophage infiltration, and twitching in murine globoid cell leukodystrophy. Cell. 1994;78(4):645–656. doi: 10.1016/0092-8674(94)90529-0.
    1. Louveau A, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523(7560):337–341. doi: 10.1038/nature14432.
    1. Ginhoux F, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330(6005):841–845. doi: 10.1126/science.1194637.
    1. Mildner A, et al. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat Neurosci. 2007;10(12):1544–1553. doi: 10.1038/nn2015.
    1. Lampron A, Pimentel-Coelho PM, Rivest S. Migration of bone marrow-derived cells into the central nervous system in models of neurodegeneration. J Comp Neurol. 2013;521(17):3863–3876.
    1. McMahon EJ, Suzuki K, Matsushima GK. Peripheral macrophage recruitment in cuprizone-induced CNS demyelination despite an intact blood-brain barrier. J Neuroimmunol. 2002;130(1–2):32–45.
    1. Wohleb ES, McKim DB, Sheridan JF, Godbout JP. Monocyte trafficking to the brain with stress and inflammation: a novel axis of immune-to-brain communication that influences mood and behavior. Front Neurosci. 2014;8:e86667.
    1. Reader BF, Jarrett BL, McKim DB, Wohleb ES, Godbout JP, Sheridan JF. Peripheral and central effects of repeated social defeat stress: monocyte trafficking, microglial activation, and anxiety. Neuroscience. 2015;289:429–442. doi: 10.1016/j.neuroscience.2015.01.001.
    1. Miron VE, Franklin RJ. Macrophages and CNS remyelination. J Neurochem. 2014;130(2):165–171. doi: 10.1111/jnc.12705.
    1. Lampron A, et al. Inefficient clearance of myelin debris by microglia impairs remyelinating processes. J Exp Med. 2015;212(4):481–495. doi: 10.1084/jem.20141656.
    1. Shechter R, et al. Recruitment of beneficial M2 macrophages to injured spinal cord is orchestrated by remote brain choroid plexus. Immunity. 2013;38(3):555–569. doi: 10.1016/j.immuni.2013.02.012.
    1. Ruckh JM, et al. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell. 2012;10(1):96–103. doi: 10.1016/j.stem.2011.11.019.
    1. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol. 2011;11(11):762–774. doi: 10.1038/nri3070.
    1. Shechter R, Schwartz M. Harnessing monocyte-derived macrophages to control central nervous system pathologies: no longer ‘if’ but ‘how’. J Pathol. 2013;229(2):332–346. doi: 10.1002/path.4106.
    1. Sanberg PR, et al. Monocyte transplantation for neural and cardiovascular ischemia repair. J Cell Mol Med. 2010;14(3):553–563.
    1. Sun JM, Kurtzberg J. Cord blood for brain injury. Cytotherapy. 2015;17(6):775–785. doi: 10.1016/j.jcyt.2015.03.004.
    1. Womble TA, et al. Monocytes are essential for the neuroprotective effect of human cord blood cells following middle cerebral artery occlusion in rat. Mol Cell Neurosci. 2014;59:76–84. doi: 10.1016/j.mcn.2014.01.004.
    1. Kurtzberg J, et al. Preclinical characterization of DUOC-01, a cell therapy product derived from banked umbilical cord blood for use as an adjuvant to umbilical cord blood transplantation for treatment of inherited metabolic diseases. Cytotherapy. 2015;17(6):803–815. doi: 10.1016/j.jcyt.2015.02.006.
    1. Zendedel A, Beyer C, Kipp M. Cuprizone-induced demyelination as a tool to study remyelination and axonal protection. J Mol Neurosci. 2013;51(2):567–572. doi: 10.1007/s12031-013-0026-4.
    1. Skripuletz T, Gudi V, Hackstette D, Stangel M. De- and remyelination in the CNS white and grey matter induced by cuprizone: the old, the new, and the unexpected. Histol Histopathol. 2011;26(12):1585–1597.
    1. Kipp M, Clarner T, Dang J, Copray S, Beyer C. The cuprizone animal model: new insights into an old story. Acta Neuropathol. 2009;118(6):723–736. doi: 10.1007/s00401-009-0591-3.
    1. Torkildsen O, Brunborg LA, Myhr KM, Bø L. The cuprizone model for demyelination. Acta Neurol Scand, Suppl. 2008;188:72–76.
    1. Matsushima GK, Morell P. The neurotoxicant, cuprizone, as a model to study demyelination and remyelination in the central nervous system. Brain Pathol. 2001;11(1):107–116.
    1. Bénardais K, et al. Cuprizone [bis(cyclohexylidenehydrazide)] is selectively toxic for mature oligodendrocytes. Neurotox Res. 2013;24(2):244–250. doi: 10.1007/s12640-013-9380-9.
    1. Einstein O, Friedman-Levi Y, Grigoriadis N, Ben-Hur T. Transplanted neural precursors enhance host brain-derived myelin regeneration. J Neurosci. 2009;29(50):15694–15702. doi: 10.1523/JNEUROSCI.3364-09.2009.
    1. Crocker SJ, et al. Intravenous administration of human embryonic stem cell-derived neural precursor cells attenuates cuprizone-induced central nervous system (CNS) demyelination. Neuropathol Appl Neurobiol. 2011;37(6):643–653. doi: 10.1111/j.1365-2990.2011.01165.x.
    1. Hedayatpour A, et al. Promotion of remyelination by adipose mesenchymal stem cell transplantation in a cuprizone model of multiple sclerosis. Cell J. 2013;15(2):142–151.
    1. Nessler J, et al. Effects of murine and human bone marrow-derived mesenchymal stem cells on cuprizone induced demyelination. PLoS One. 2013;8(7):e86667. doi: 10.1371/journal.pone.0069795.
    1. Arnett HA, et al. bHLH transcription factor Olig1 is required to repair demyelinated lesions in the CNS. Science. 2004;306(5704):2111–2115. doi: 10.1126/science.1103709.
    1. Mason JL, et al. Oligodendrocytes and progenitors become progressively depleted within chronically demyelinated lesions. Am J Pathol. 2004;164(5):1673–1682. doi: 10.1016/S0002-9440(10)63726-1.
    1. Gudi V, Gingele S, Skripuletz T, Stangel M. Glial response during cuprizone-induced de- and remyelination in the CNS: lessons learned. Front Cell Neurosci. 2014;8:e86667.
    1. Mason JL, Ye P, Suzuki K, D’Ercole AJ, Matsushima GK. Insulin-like growth factor-1 inhibits mature oligodendrocyte apoptosis during primary demyelination. J Neurosci. 2000;20(15):5703–5708.
    1. Biancotti JC, Kumar S, de Vellis J. Activation of inflammatory response by a combination of growth factors in cuprizone-induced demyelinated brain leads to myelin repair. Neurochem Res. 2008;33(12):2615–2628. doi: 10.1007/s11064-008-9792-8.
    1. Skripuletz T, et al. Astrocytes regulate myelin clearance through recruitment of microglia during cuprizone-induced demyelination. Brain. 2013;136(Pt 1):147–167.
    1. Clarner T, et al. Myelin debris regulates inflammatory responses in an experimental demyelination animal model and multiple sclerosis lesions. Glia. 2012;60(10):1468–1480. doi: 10.1002/glia.22367.
    1. Hiremath MM, Saito Y, Knapp GW, Ting JP, Suzuki K, Matsushima GK. Microglial/macrophage accumulation during cuprizone-induced demyelination in C57BL/6 mice. J Neuroimmunol. 1998;92(1–2):38–49.
    1. Hibbits N, Yoshino J, Le TQ, Armstrong RC. Astrogliosis during acute and chronic cuprizone demyelination and implications for remyelination. ASN Neuro. 2012;4(6):393–408. doi: 10.1042/AN20120062.
    1. Shultz LD, et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells. J Immunol. 2005;174(10):6477–6489. doi: 10.4049/jimmunol.174.10.6477.
    1. Praet J, Guglielmetti C, Berneman Z, Van der Linden A, Ponsaerts P. Cellular and molecular neuropathology of the cuprizone mouse model: clinical relevance for multiple sclerosis. Neurosci Biobehav Rev. 2014;47:485–505. doi: 10.1016/j.neubiorev.2014.10.004.
    1. Karrer FM, Reitz BL, Hao L, Lafferty KJ. Fluorescein labeling of murine hepatocytes for identification after intrahepatic transplantation. Transplant Proc. 1992;24(6):2820–2821.
    1. Dimou L, Simon C, Kirchhoff F, Takebayashi H, Götz M. Progeny of Olig2-expressing progenitors in the gray and white matter of the adult mouse cerebral cortex. J Neurosci. 2008;28(41):10434–10442. doi: 10.1523/JNEUROSCI.2831-08.2008.
    1. Fancy SP, Chan JR, Baranzini SE, Franklin RJ, Rowitch DH. Myelin regeneration: a recapitulation of development? Annu Rev Neurosci. 2011;34:21–43. doi: 10.1146/annurev-neuro-061010-113629.
    1. Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown. J Cell Physiol. 2000;182(3):311–322. doi: 10.1002/(SICI)1097-4652(200003)182:3<311::AID-JCP1>;2-9.
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57.
    1. Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37(1):1–13. doi: 10.1093/nar/gkn923.
    1. Misumi Y, Misumi Y, Miki K, Takatsuki A, Tamura G, Ikehara Y. Novel blockade by brefeldin A of intracellular transport of secretory proteins in cultured rat hepatocytes. J Biol Chem. 1986;261(24):11398–11403.
    1. Storms R, et al. Tissue distribution of a cord blood-derived cell product following intrathecal transplantation. Cytotherapy. 2014;16(4):e86667
    1. Kashani IR, et al. Protective effects of melatonin against mitochondrial injury in a mouse model of multiple sclerosis. Exp Brain Res. 2014;232(9):2835–2846. doi: 10.1007/s00221-014-3946-5.
    1. Tandler B, Hoppel CL. Division of giant mitochondria during recovery from cuprizone intoxication. J Cell Biol. 1973;56(1):266–272. doi: 10.1083/jcb.56.1.266.
    1. Acs P, Komoly S. Selective ultrastructural vulnerability in the cuprizone-induced experimental demyelination. Ideggyogy Sz. 2012;65(7–8):266–270.
    1. Flatmark T, Kryvi H, Tangerås A. Induction of megamitochondria by cuprizone(biscyclohexanone oxaldihydrazone). Evidence for an inhibition of the mitochondrial division process. Eur J Cell Biol. 1980;23(1):141–148.
    1. Asano M, Wakabayashi T, Ishikawa K, Kishimoto H. Mechanism of the formation of megamitochondria by copper-chelating agents. IV. Role of fusion phenomenon in the cuprizone-induced megamitochondrial formation. Acta Pathol Jpn. 1978;28(2):205–213.
    1. Woodruff RH, Fruttiger M, Richardson WD, Franklin RJ. Platelet-derived growth factor regulates oligodendrocyte progenitor numbers in adult CNS and their response following CNS demyelination. Mol Cell Neurosci. 2004;25(2):252–262. doi: 10.1016/j.mcn.2003.10.014.
    1. Murtie JC, Zhou YX, Le TQ, Vana AC, Armstrong RC. PDGF and FGF2 pathways regulate distinct oligodendrocyte lineage responses in experimental demyelination with spontaneous remyelination. Neurobiol Dis. 2005;19(1–2):171–182.
    1. Vana AC, Flint NC, Harwood NE, Le TQ, Fruttiger M, Armstrong RC. Platelet-derived growth factor promotes repair of chronically demyelinated white matter. J Neuropathol Exp Neurol. 2007;66(11):975–988. doi: 10.1097/NEN.0b013e3181587d46.
    1. Ida JA, Dubois-Dalcq M, McKinnon RD. Expression of the receptor tyrosine kinase c-kit in oligodendrocyte progenitor cells. J Neurosci Res. 1993;36(5):596–606. doi: 10.1002/jnr.490360512.
    1. Erlandsson A, Larsson J, Forsberg-Nilsson K. Stem cell factor is a chemoattractant and a survival factor for CNS stem cells. Exp Cell Res. 2004;301(2):201–210. doi: 10.1016/j.yexcr.2004.08.009.
    1. D’Ercole AJ, Ye P, Calikoglu AS, Gutierrez-Ospina G. The role of the insulin-like growth factors in the central nervous system. Mol Neurobiol. 1996;13(3):227–255. doi: 10.1007/BF02740625.
    1. García-Segura LM, et al. Interaction of the signalling pathways of insulin-like growth factor-I and sex steroids in the neuroendocrine hypothalamus. Horm Res. 1996;46(4–5):160–164.
    1. Cho KH, Kim MW, Kim SU. Tissue culture model of Krabbe’s disease: psychosine cytotoxicity in rat oligodendrocyte culture. Dev Neurosci. 1997;19(4):321–327.
    1. Wang Y, et al. TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell. 2015;160(6):1061–1071. doi: 10.1016/j.cell.2015.01.049.
    1. Cantoni C, et al. TREM2 regulates microglial cell activation in response to demyelination in vivo. Acta Neuropathol. 2015;129(3):429–447. doi: 10.1007/s00401-015-1388-1.
    1. Tsiperson V, Li X, Schwartz GJ, Raine CS, Shafit-Zagardo B. GAS6 enhances repair following cuprizone-induced demyelination. PLoS One. 2010;5(12):e86667. doi: 10.1371/journal.pone.0015748.
    1. Butovsky O, et al. Microglia activated by IL-4 or IFN-gamma differentially induce neurogenesis and oligodendrogenesis from adult stem/progenitor cells. Mol Cell Neurosci. 2006;31(1):149–160. doi: 10.1016/j.mcn.2005.10.006.
    1. Yang J, et al. Adult neural stem cells expressing IL-10 confer potent immunomodulation and remyelination in experimental autoimmune encephalitis. J Clin Invest. 2009;119(12):3678–3691. doi: 10.1172/JCI37914.
    1. Strle K, et al. Interleukin-10 in the brain. Crit Rev Immunol. 2001;21(5):427–449.
    1. Molina-Holgado F, Grencis R, Rothwell NJ. Actions of exogenous and endogenous IL-10 on glial responses to bacterial LPS/cytokines. Glia. 2001;33(2):97–106.
    1. Boyd ZS, Kriatchko A, Yang J, Agarwal N, Wax MB, Patil RV. Interleukin-10 receptor signaling through STAT-3 regulates the apoptosis of retinal ganglion cells in response to stress. Invest Ophthalmol Vis Sci. 2003;44(12):5206–5211. doi: 10.1167/iovs.03-0534.
    1. Arnett HA, Mason J, Marino M, Suzuki K, Matsushima GK, Ting JP. TNF alpha promotes proliferation of oligodendrocyte progenitors and remyelination. Nat Neurosci. 2001;4(11):1116–1122. doi: 10.1038/nn738.
    1. Larsen PH, Wells JE, Stallcup WB, Opdenakker G, Yong VW. Matrix metalloproteinase-9 facilitates remyelination in part by processing the inhibitory NG2 proteoglycan. J Neurosci. 2003;23(35):11127–11135.
    1. Whitelock JM, Murdoch AD, Iozzo RV, Underwood PA. The degradation of human endothelial cell-derived perlecan and release of bound basic fibroblast growth factor by stromelysin, collagenase, plasmin, and heparanases. J Biol Chem. 1996;271(17):10079–10086. doi: 10.1074/jbc.271.17.10079.
    1. Yong VW, Power C, Forsyth P, Edwards DR. Metalloproteinases in biology and pathology of the nervous system. Nat Rev Neurosci. 2001;2(7):502–511. doi: 10.1038/35081571.
    1. Doan V, Kleindienst AM, McMahon EJ, Long BR, Matsushima GK, Taylor LC. Abbreviated exposure to cuprizone is sufficient to induce demyelination and oligodendrocyte loss. J Neurosci Res. 2013;91(3):363–373. doi: 10.1002/jnr.23174.
    1. Goebbels S, et al. Elevated phosphatidylinositol 3,4,5-trisphosphate in glia triggers cell-autonomous membrane wrapping and myelination. J Neurosci. 2010;30(26):8953–8964. doi: 10.1523/JNEUROSCI.0219-10.2010.

Source: PubMed

3
Iratkozz fel