Early intervention for spinal cord injury with human induced pluripotent stem cells oligodendrocyte progenitors

Angelo H All, Payam Gharibani, Siddharth Gupta, Faith A Bazley, Nikta Pashai, Bin-Kuan Chou, Sandeep Shah, Linda M Resar, Linzhao Cheng, John D Gearhart, Candace L Kerr, Angelo H All, Payam Gharibani, Siddharth Gupta, Faith A Bazley, Nikta Pashai, Bin-Kuan Chou, Sandeep Shah, Linda M Resar, Linzhao Cheng, John D Gearhart, Candace L Kerr

Abstract

Induced pluripotent stem (iPS) cells are at the forefront of research in regenerative medicine and are envisaged as a source for personalized tissue repair and cell replacement therapy. Here, we demonstrate for the first time that oligodendrocyte progenitors (OPs) can be derived from iPS cells generated using either an episomal, non-integrating plasmid approach or standard integrating retroviruses that survive and differentiate into mature oligodendrocytes after early transplantation into the injured spinal cord. The efficiency of OP differentiation in all 3 lines tested ranged from 40% to 60% of total cells, comparable to those derived from human embryonic stem cells. iPS cell lines derived using episomal vectors or retroviruses generated a similar number of early neural progenitors and glial progenitors while the episomal plasmid-derived iPS line generated more OPs expressing late markers O1 and RIP. Moreover, we discovered that iPS-derived OPs (iPS-OPs) engrafted 24 hours following a moderate contusive spinal cord injury (SCI) in rats survived for approximately two months and that more than 70% of the transplanted cells differentiated into mature oligodendrocytes that expressed myelin associated proteins. Transplanted OPs resulted in a significant increase in the number of myelinated axons in animals that received a transplantation 24 h after injury. In addition, nearly a 5-fold reduction in cavity size and reduced glial scarring was seen in iPS-treated groups compared to the control group, which was injected with heat-killed iPS-OPs. Although further investigation is needed to understand the mechanisms involved, these results provide evidence that patient-specific, iPS-derived OPs can survive for three months and improve behavioral assessment (BBB) after acute transplantation into SCI. This is significant as determining the time in which stem cells are injected after SCI may influence their survival and differentiation capacity.

Conflict of interest statement

Competing Interests: LC is a PLOS ONE Editorial Board member. This does not alter the authors' adherence to PLOS ONE Editorial policies and criteria.

Figures

Fig 1. Bright field microscopy images of…
Fig 1. Bright field microscopy images of the various stages of oligodendrocyte differentiation from induced pluripotent stem cells of the BC1 line.
(A) The undifferentiated cells grew in colonies on a mouse embryonic fibroblast feeder layer, (B) Embroid bodies (EBs) were formed by suspending the undifferentiating cells in culture on ultra-low adherence plates, (C) 15-day EBs were plated on matrigel, leading to the extension of cells of neuroectodermal lineage. Upon passaging, these neural progenitors grew in a monolayer and were subsequently differentiated to glial progenitors (D) stage. Addition of platelet derived growth factor-AA (PDGF-AA) to the media led to glial progenitors (GPs) transitioning into the (E) oligodendrocyte progenitor (OP) stage, displaying a rounded morphology with multiple filopodial extensions. (F) Terminal differentiation was induced by the removal of PDGF-AA, leading to the appearance of mature oligodendrocytes displaying characteristic branching. Scale bars- A-B: 300 μm, C-F: 80 μm
Fig 2. Plots showing mRNA expression of…
Fig 2. Plots showing mRNA expression of various developmental markers for all three iPS lines at each stage ofoligodendrocyte differentiation, as measured using qRT-PCR and normalized to fetal fibroblast controls.
Expression of pluripotency associated genes (A) NANOG, (B) Octamer-binding transcription factor 4 (OCT4) also known as POU5F1 (POU domain, class 5, transcription factor 1) and (C) SRY (sex determining region Y)-box 2 (SOX2) were undetected in oligodendrocyte progenitors (OPs) whilemarkers indicative of the oligodendrocyte lineage were induced including (D) NG2, (E) Myelin-associated glycoprotein (MAG), and (F) Myelin-associated oligodendrocytic basic protein (MOBP). Delta CTs were calculated using β-actin as a relative standard and lines normalized to fetal fibroblast controls. Mean + SEM were calculated from two independent differentiation experiments for each line and each gene performed in technical triplicate.Positive control included total RNA from human fetal brain. Abbreviations are Undiff: Undifferentiated iPS cells, EB: embryoid body, NP: neural progenitor, GP: glial progenitor, OP: oligodendrocyte progenitor.
Fig 3. Indirect immunofluorescence detection of neural…
Fig 3. Indirect immunofluorescence detection of neural markers help define the initial stages of oligodendrocyte differentiation from undifferentiated iPS cells, into neural (NP) and glial (GP) progenitors.
(A-D) Photos are of cells derived from BC1 with the differentiated stage (underlined) and marker analyzed as indicated similar staining patterns were seen in all three lines. Nuclei were stained using DAPI (blue). Most neural progenitors (NPs) were (A) NESTIN+ and (B) A2B5+ while the majority of glial progenitors (GPs) began expressing early oligodendrocyte progenitor (OP) markers (C) platelet derived growth factor receptor-α (PDGFR-α) and (D) NG2. Lower panel shows quantification of the indirect immunofluorescence analyses calculating the percent of labeled cells in all markers. (E) 90% of all cells from each line expressed early markers of the neural lineage including NESTIN and A2B5. (F) Likewise the number of glial progenitors expressing markers of the early OP lineage such as PDGFR-α and NG2 were also similar among lines. Differentiation of each cell line was performed in triplicate and the percent of positively stained cells was determined from 3 randomly, chosen 10X fields per 24-well cell culture dish from all three replicates. Values in the graph represent mean ± SEM. * and † denote statistical significance compared to BC1 and MR31, respectively by ANOVA and Tukey post hoc tests (N = 9, P

Fig 4. Indirect immunofluorescence marker characterization of…

Fig 4. Indirect immunofluorescence marker characterization of oligodendrocyte progenitors derived from human iPS cells.

Nuclei…

Fig 4. Indirect immunofluorescence marker characterization of oligodendrocyte progenitors derived from human iPS cells.
Nuclei were stained with DAPI (blue). Results show reduced expression of early markers (A) A2B5 and (B) NG2 and increased protein expression of mid to late oligodendrocyte progenitor (OP) differentiation markers (C) O4 (D) RIP (E) O1 (F) myelin-associated glycoprotein (MAG) and (G) myelin-associated oligodendrocytic basic protein (MOBP). Myelin basic protein (MBP) expression was not detectable by immunostaining. 1% or less of OP cells expressed markers of other neural lineages as denoted by (H) the neuronal maker neuron-specific class III beta-tubulin (TUJ1) and (I) the astrocyte marker glial fibrillary acidic protein (GFAP). (J) Quantification of the indirect immunofluorescence analyses calculating the percent of cells expressing a particular marker is shown in the lower panel. A higher percent of cells derived from BC1 expressed later OP markers MAG, MOBP and O1 while fewer cells from the BC1 lines expressed early neural progenitor (NP) and glial progenitor (GP) markers and early OP marker O4 compared to MR31 and A1-4. Differentiation of each cell line was performed in triplicate and the percent of positively stained cells was determined from 3 randomly, chosen 10X fields per 24-well cell culture dish from all three replicates. Values in the graphs represent mean ± SEM. * and † denote statistical significance compared to BC1 and MR31, respectively by ANOVA and Tukey post hoc tests (N = 9, P

Fig 5. Immunohistological characterization in rat spinal…

Fig 5. Immunohistological characterization in rat spinal cords at the epicenter of injury (asterisks) following…

Fig 5. Immunohistological characterization in rat spinal cords at the epicenter of injury (asterisks) following transplantation of either (A) heat-killed iPS-OPs or (B) live iPS-OPs, 24 hours after injury.
Human specific anti-nuclei antibody (HNA) staining showed that cells transplanted at the site of injury could survive, differentiate and express myelin basic protein (MBP, red) in iPS-OPs group, compared to the heat-killed iPS-OPs group. (C) Heat-killed iPS-OPs treated spinal cords demonstrated more cavitation and higher glial fibrillary acidic protein (GFAP) (green) expression than (D) spinal cords treated with iPS-OPs. Heat-killed iPS-OPs also expressed less MBP, a mature oligodendrocyte marker, than live iPS-OPs (red). (E) Double immunostaining with CD68 (expressed by macrophages, microglial, and T cells) and MBP shows more CD68+/MBP+ cells in control animals with morphology indicative of active macrophages or microglial engulfment of OPs. (F) Spinal cords injected with iPS-OPs had significantly fewer CD68+/MBP+ cells. Tissues shown here were isolated approximately 2 months after transplantation. DAPI was used to stain rat and human nuclei (blue). Scale bars for A, C, D: 100 μm and B, E, F: 30 μm. (G) Quantitative image analysis shows that more than 70% of HNA+ cells were differentiated MBP+ oligodendrocytes. Only 4.4 ± 1.5% of grafted cells express glial fibrillary acidic protein (GFAP), while no HNA+ cells were detected with co-localized staining for neuronal marker neuron-specific class III beta-tubulin (TUJ1) or NESTIN. Values represent mean ± SEM. * denote statistical significance of human MBP+ oligodendrocytes and MBP- OPs compared to either human-derived neural progenitors (Nestin+), neurons (TUJ1+) or astrocytes (GFAP+) and † denotes statistical significance of human HNA+MBP+ compared to HNA+MBP- OPs by Tukey post hoc tests. Tissue slices encompassing the injury epicenter were stained in intervals for each antibody combination from 4 rats in each treatment group, P

Fig 6. Hematoxylin and eosin and luxol…

Fig 6. Hematoxylin and eosin and luxol fast blue stained paraffin sections of spinal cords…

Fig 6. Hematoxylin and eosin and luxol fast blue stained paraffin sections of spinal cords from animals subjected to moderate contusive SCI and treated with (A) Heat-killed iPS-OPs or (B) Live, intact iPS-OPs derived from the BC1 line.
As expected for a 12.5 mm contusive injury, cavity formation was seen in both groups; however, the cavitation in spinal cords treated with iPS-OPs did not form well defined glial-like scar structures around the cavity periphery, compared to the control group. Thus, controls showed more pronounced cavitation, which extended further from the epicenter along the central canal. This indicates that the transplantation of live iPS-OPs into the newly formed cavity 24 hours after injury limited the normal expected progression of this level of contusive injury. The asterisks indicate the center of the injury site. Yellow boxes in A and B correspond to adjacent tissue sections stained in C and D, respectively. In these areas immediately surrounding the center of injury, staining with Luxol Fast Blue (LFB) to identify myelin showed reduced staining in the control group (C) versus the iPS-OP transplanted spinal cords (D). Thus, higher levels of LFB in the iPS-OP treated rats may indicate the presence of greater amounts of myelin. The representative spinal cords shown in (A-D) were harvested approximately 2 months after transplantation. Dotted lines help delineate regions of white matter (W) and grey matter (G). (E) Area measurements across groups revealed nearly a 5-fold significant reduction in cavity size in the iPS-OP treated groups compared to the heat-killed controls. Optical density of LBF, normalized to the optical density of the sham control group, was calculated for the grey matter (F) and white matter (G) of spinal sections to quantify the amount of myelin present in each area of the cord. Values in the graphs represent mean ± SEM. * denote statistical significance compared to heat-killed iPS-OPs by Tukey post hoc tests. For each stain, 5 identically numbered tissue slices encompassing the injury epicenter from 4 rats were analyzed for each treatment group, P

Fig 7. Transmission electron microscopy of transverse…

Fig 7. Transmission electron microscopy of transverse sections of the spinal cord showed remyelination of…

Fig 7. Transmission electron microscopy of transverse sections of the spinal cord showed remyelination of spared axons by BC1-derived iPS-OP cells after contusive SCI in rats.
(A) Normal, endogenous myelination of an uninjured rat spinal cord (laminectomy sham control) as indicated by white arrows, showing thick and tightly-packed myelin sheaths. (B) Remyelination observed in rats transplanted with BC1-derived OPs, approximately two months after transplantation. These are sites where oligodendrocytes are detected, as shown in Fig. 4. The relatively thinner and loosely packed myelin sheaths may be compared with the normal condition in (A), suggesting that this is remyelination by human OPs. (C) Multiple nude axons are visible in rats injected with heat-killed iPS-OPs, with no observable remyelination, approximately two months after transplant. (D) Quantification shows that transplantation of BC1-derived OPs resulted in a significant increase in the number of remyelinated axons and reduced demyelinated of axons compared with the heat-killed iPS-OP group. Values in the graphs represent mean ± SEM. * denote statistical significance compared to heat-killed iPS-OPs by Tukey post hoc tests. Ten images encompassing the injury epicenter from 4 rats were analyzed for each treatment group (P

Fig 8. Behavioral assessment 8 weeks following…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in rats.
The average of the Basso, Beattie, and Bresnahan (BBB) Locomotor Rating Scale (BBB SCORE) for left and right limb movement was evaluated for 55 days following spinal cord injury in rats injected with either PBS (blue), 500,000 heat-killed iPS-OP cells (green), live BC1-OP (black), or MR31-OP (red) cells 24 hrs after injury. Values in the graphs represent mean ± SEM of both left and right limbs as there were no significant differences in response between right and left limb scores. Statistical significance is denoted by a colored asterisk for each cell line with matched color compared to the PBS control group determined by one-way ANOVA followed by a two-way Student’s t-test assuming equal variances (P
All figures (8)
Similar articles
Cited by
References
    1. Gunaseeli I, Doss MX, Antzelevitch C, Hescheler J, Sachinidis A (2010) Induced pluripotent stem cells as a model for accelerated patient- and disease-specific drug discovery. Curr Med Chem 17: 759–766. - PMC - PubMed
    1. Lau F, Ahfeldt T, Osafune K, Akustsu H, Cowan CA (2009) Induced pluripotent stem (iPS) cells: an up-to-the-minute review. F1000 Biol Rep 1: 84 10.3410/B1-84 - DOI - PMC - PubMed
    1. Nishikawa S, Goldstein RA, Nierras CR (2008) The promise of human induced pluripotent stem cells for research and therapy. Nat Rev Mol Cell Biol 9: 725–729. 10.1038/nrm2466 - DOI - PubMed
    1. Wang S, Bates J, Li X, Schanz S, Chandler-Militello D, et al. (2013) Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 12: 252–264. 10.1016/j.stem.2012.12.002 - DOI - PMC - PubMed
    1. Yoshihara H, Shumsky JS, Neuhuber B, Otsuka T, Fischer I, et al. (2006) Combining motor training with transplantation of rat bone marrow stromal cells does not improve repair or recovery in rats with thoracic contusion injuries. Brain Research 1119: 65–75. - PubMed
Show all 78 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig 4. Indirect immunofluorescence marker characterization of…
Fig 4. Indirect immunofluorescence marker characterization of oligodendrocyte progenitors derived from human iPS cells.
Nuclei were stained with DAPI (blue). Results show reduced expression of early markers (A) A2B5 and (B) NG2 and increased protein expression of mid to late oligodendrocyte progenitor (OP) differentiation markers (C) O4 (D) RIP (E) O1 (F) myelin-associated glycoprotein (MAG) and (G) myelin-associated oligodendrocytic basic protein (MOBP). Myelin basic protein (MBP) expression was not detectable by immunostaining. 1% or less of OP cells expressed markers of other neural lineages as denoted by (H) the neuronal maker neuron-specific class III beta-tubulin (TUJ1) and (I) the astrocyte marker glial fibrillary acidic protein (GFAP). (J) Quantification of the indirect immunofluorescence analyses calculating the percent of cells expressing a particular marker is shown in the lower panel. A higher percent of cells derived from BC1 expressed later OP markers MAG, MOBP and O1 while fewer cells from the BC1 lines expressed early neural progenitor (NP) and glial progenitor (GP) markers and early OP marker O4 compared to MR31 and A1-4. Differentiation of each cell line was performed in triplicate and the percent of positively stained cells was determined from 3 randomly, chosen 10X fields per 24-well cell culture dish from all three replicates. Values in the graphs represent mean ± SEM. * and † denote statistical significance compared to BC1 and MR31, respectively by ANOVA and Tukey post hoc tests (N = 9, P

Fig 5. Immunohistological characterization in rat spinal…

Fig 5. Immunohistological characterization in rat spinal cords at the epicenter of injury (asterisks) following…

Fig 5. Immunohistological characterization in rat spinal cords at the epicenter of injury (asterisks) following transplantation of either (A) heat-killed iPS-OPs or (B) live iPS-OPs, 24 hours after injury.
Human specific anti-nuclei antibody (HNA) staining showed that cells transplanted at the site of injury could survive, differentiate and express myelin basic protein (MBP, red) in iPS-OPs group, compared to the heat-killed iPS-OPs group. (C) Heat-killed iPS-OPs treated spinal cords demonstrated more cavitation and higher glial fibrillary acidic protein (GFAP) (green) expression than (D) spinal cords treated with iPS-OPs. Heat-killed iPS-OPs also expressed less MBP, a mature oligodendrocyte marker, than live iPS-OPs (red). (E) Double immunostaining with CD68 (expressed by macrophages, microglial, and T cells) and MBP shows more CD68+/MBP+ cells in control animals with morphology indicative of active macrophages or microglial engulfment of OPs. (F) Spinal cords injected with iPS-OPs had significantly fewer CD68+/MBP+ cells. Tissues shown here were isolated approximately 2 months after transplantation. DAPI was used to stain rat and human nuclei (blue). Scale bars for A, C, D: 100 μm and B, E, F: 30 μm. (G) Quantitative image analysis shows that more than 70% of HNA+ cells were differentiated MBP+ oligodendrocytes. Only 4.4 ± 1.5% of grafted cells express glial fibrillary acidic protein (GFAP), while no HNA+ cells were detected with co-localized staining for neuronal marker neuron-specific class III beta-tubulin (TUJ1) or NESTIN. Values represent mean ± SEM. * denote statistical significance of human MBP+ oligodendrocytes and MBP- OPs compared to either human-derived neural progenitors (Nestin+), neurons (TUJ1+) or astrocytes (GFAP+) and † denotes statistical significance of human HNA+MBP+ compared to HNA+MBP- OPs by Tukey post hoc tests. Tissue slices encompassing the injury epicenter were stained in intervals for each antibody combination from 4 rats in each treatment group, P

Fig 6. Hematoxylin and eosin and luxol…

Fig 6. Hematoxylin and eosin and luxol fast blue stained paraffin sections of spinal cords…

Fig 6. Hematoxylin and eosin and luxol fast blue stained paraffin sections of spinal cords from animals subjected to moderate contusive SCI and treated with (A) Heat-killed iPS-OPs or (B) Live, intact iPS-OPs derived from the BC1 line.
As expected for a 12.5 mm contusive injury, cavity formation was seen in both groups; however, the cavitation in spinal cords treated with iPS-OPs did not form well defined glial-like scar structures around the cavity periphery, compared to the control group. Thus, controls showed more pronounced cavitation, which extended further from the epicenter along the central canal. This indicates that the transplantation of live iPS-OPs into the newly formed cavity 24 hours after injury limited the normal expected progression of this level of contusive injury. The asterisks indicate the center of the injury site. Yellow boxes in A and B correspond to adjacent tissue sections stained in C and D, respectively. In these areas immediately surrounding the center of injury, staining with Luxol Fast Blue (LFB) to identify myelin showed reduced staining in the control group (C) versus the iPS-OP transplanted spinal cords (D). Thus, higher levels of LFB in the iPS-OP treated rats may indicate the presence of greater amounts of myelin. The representative spinal cords shown in (A-D) were harvested approximately 2 months after transplantation. Dotted lines help delineate regions of white matter (W) and grey matter (G). (E) Area measurements across groups revealed nearly a 5-fold significant reduction in cavity size in the iPS-OP treated groups compared to the heat-killed controls. Optical density of LBF, normalized to the optical density of the sham control group, was calculated for the grey matter (F) and white matter (G) of spinal sections to quantify the amount of myelin present in each area of the cord. Values in the graphs represent mean ± SEM. * denote statistical significance compared to heat-killed iPS-OPs by Tukey post hoc tests. For each stain, 5 identically numbered tissue slices encompassing the injury epicenter from 4 rats were analyzed for each treatment group, P

Fig 7. Transmission electron microscopy of transverse…

Fig 7. Transmission electron microscopy of transverse sections of the spinal cord showed remyelination of…

Fig 7. Transmission electron microscopy of transverse sections of the spinal cord showed remyelination of spared axons by BC1-derived iPS-OP cells after contusive SCI in rats.
(A) Normal, endogenous myelination of an uninjured rat spinal cord (laminectomy sham control) as indicated by white arrows, showing thick and tightly-packed myelin sheaths. (B) Remyelination observed in rats transplanted with BC1-derived OPs, approximately two months after transplantation. These are sites where oligodendrocytes are detected, as shown in Fig. 4. The relatively thinner and loosely packed myelin sheaths may be compared with the normal condition in (A), suggesting that this is remyelination by human OPs. (C) Multiple nude axons are visible in rats injected with heat-killed iPS-OPs, with no observable remyelination, approximately two months after transplant. (D) Quantification shows that transplantation of BC1-derived OPs resulted in a significant increase in the number of remyelinated axons and reduced demyelinated of axons compared with the heat-killed iPS-OP group. Values in the graphs represent mean ± SEM. * denote statistical significance compared to heat-killed iPS-OPs by Tukey post hoc tests. Ten images encompassing the injury epicenter from 4 rats were analyzed for each treatment group (P

Fig 8. Behavioral assessment 8 weeks following…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in rats.
The average of the Basso, Beattie, and Bresnahan (BBB) Locomotor Rating Scale (BBB SCORE) for left and right limb movement was evaluated for 55 days following spinal cord injury in rats injected with either PBS (blue), 500,000 heat-killed iPS-OP cells (green), live BC1-OP (black), or MR31-OP (red) cells 24 hrs after injury. Values in the graphs represent mean ± SEM of both left and right limbs as there were no significant differences in response between right and left limb scores. Statistical significance is denoted by a colored asterisk for each cell line with matched color compared to the PBS control group determined by one-way ANOVA followed by a two-way Student’s t-test assuming equal variances (P
All figures (8)
Similar articles
Cited by
References
    1. Gunaseeli I, Doss MX, Antzelevitch C, Hescheler J, Sachinidis A (2010) Induced pluripotent stem cells as a model for accelerated patient- and disease-specific drug discovery. Curr Med Chem 17: 759–766. - PMC - PubMed
    1. Lau F, Ahfeldt T, Osafune K, Akustsu H, Cowan CA (2009) Induced pluripotent stem (iPS) cells: an up-to-the-minute review. F1000 Biol Rep 1: 84 10.3410/B1-84 - DOI - PMC - PubMed
    1. Nishikawa S, Goldstein RA, Nierras CR (2008) The promise of human induced pluripotent stem cells for research and therapy. Nat Rev Mol Cell Biol 9: 725–729. 10.1038/nrm2466 - DOI - PubMed
    1. Wang S, Bates J, Li X, Schanz S, Chandler-Militello D, et al. (2013) Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 12: 252–264. 10.1016/j.stem.2012.12.002 - DOI - PMC - PubMed
    1. Yoshihara H, Shumsky JS, Neuhuber B, Otsuka T, Fischer I, et al. (2006) Combining motor training with transplantation of rat bone marrow stromal cells does not improve repair or recovery in rats with thoracic contusion injuries. Brain Research 1119: 65–75. - PubMed
Show all 78 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig 5. Immunohistological characterization in rat spinal…
Fig 5. Immunohistological characterization in rat spinal cords at the epicenter of injury (asterisks) following transplantation of either (A) heat-killed iPS-OPs or (B) live iPS-OPs, 24 hours after injury.
Human specific anti-nuclei antibody (HNA) staining showed that cells transplanted at the site of injury could survive, differentiate and express myelin basic protein (MBP, red) in iPS-OPs group, compared to the heat-killed iPS-OPs group. (C) Heat-killed iPS-OPs treated spinal cords demonstrated more cavitation and higher glial fibrillary acidic protein (GFAP) (green) expression than (D) spinal cords treated with iPS-OPs. Heat-killed iPS-OPs also expressed less MBP, a mature oligodendrocyte marker, than live iPS-OPs (red). (E) Double immunostaining with CD68 (expressed by macrophages, microglial, and T cells) and MBP shows more CD68+/MBP+ cells in control animals with morphology indicative of active macrophages or microglial engulfment of OPs. (F) Spinal cords injected with iPS-OPs had significantly fewer CD68+/MBP+ cells. Tissues shown here were isolated approximately 2 months after transplantation. DAPI was used to stain rat and human nuclei (blue). Scale bars for A, C, D: 100 μm and B, E, F: 30 μm. (G) Quantitative image analysis shows that more than 70% of HNA+ cells were differentiated MBP+ oligodendrocytes. Only 4.4 ± 1.5% of grafted cells express glial fibrillary acidic protein (GFAP), while no HNA+ cells were detected with co-localized staining for neuronal marker neuron-specific class III beta-tubulin (TUJ1) or NESTIN. Values represent mean ± SEM. * denote statistical significance of human MBP+ oligodendrocytes and MBP- OPs compared to either human-derived neural progenitors (Nestin+), neurons (TUJ1+) or astrocytes (GFAP+) and † denotes statistical significance of human HNA+MBP+ compared to HNA+MBP- OPs by Tukey post hoc tests. Tissue slices encompassing the injury epicenter were stained in intervals for each antibody combination from 4 rats in each treatment group, P

Fig 6. Hematoxylin and eosin and luxol…

Fig 6. Hematoxylin and eosin and luxol fast blue stained paraffin sections of spinal cords…

Fig 6. Hematoxylin and eosin and luxol fast blue stained paraffin sections of spinal cords from animals subjected to moderate contusive SCI and treated with (A) Heat-killed iPS-OPs or (B) Live, intact iPS-OPs derived from the BC1 line.
As expected for a 12.5 mm contusive injury, cavity formation was seen in both groups; however, the cavitation in spinal cords treated with iPS-OPs did not form well defined glial-like scar structures around the cavity periphery, compared to the control group. Thus, controls showed more pronounced cavitation, which extended further from the epicenter along the central canal. This indicates that the transplantation of live iPS-OPs into the newly formed cavity 24 hours after injury limited the normal expected progression of this level of contusive injury. The asterisks indicate the center of the injury site. Yellow boxes in A and B correspond to adjacent tissue sections stained in C and D, respectively. In these areas immediately surrounding the center of injury, staining with Luxol Fast Blue (LFB) to identify myelin showed reduced staining in the control group (C) versus the iPS-OP transplanted spinal cords (D). Thus, higher levels of LFB in the iPS-OP treated rats may indicate the presence of greater amounts of myelin. The representative spinal cords shown in (A-D) were harvested approximately 2 months after transplantation. Dotted lines help delineate regions of white matter (W) and grey matter (G). (E) Area measurements across groups revealed nearly a 5-fold significant reduction in cavity size in the iPS-OP treated groups compared to the heat-killed controls. Optical density of LBF, normalized to the optical density of the sham control group, was calculated for the grey matter (F) and white matter (G) of spinal sections to quantify the amount of myelin present in each area of the cord. Values in the graphs represent mean ± SEM. * denote statistical significance compared to heat-killed iPS-OPs by Tukey post hoc tests. For each stain, 5 identically numbered tissue slices encompassing the injury epicenter from 4 rats were analyzed for each treatment group, P

Fig 7. Transmission electron microscopy of transverse…

Fig 7. Transmission electron microscopy of transverse sections of the spinal cord showed remyelination of…

Fig 7. Transmission electron microscopy of transverse sections of the spinal cord showed remyelination of spared axons by BC1-derived iPS-OP cells after contusive SCI in rats.
(A) Normal, endogenous myelination of an uninjured rat spinal cord (laminectomy sham control) as indicated by white arrows, showing thick and tightly-packed myelin sheaths. (B) Remyelination observed in rats transplanted with BC1-derived OPs, approximately two months after transplantation. These are sites where oligodendrocytes are detected, as shown in Fig. 4. The relatively thinner and loosely packed myelin sheaths may be compared with the normal condition in (A), suggesting that this is remyelination by human OPs. (C) Multiple nude axons are visible in rats injected with heat-killed iPS-OPs, with no observable remyelination, approximately two months after transplant. (D) Quantification shows that transplantation of BC1-derived OPs resulted in a significant increase in the number of remyelinated axons and reduced demyelinated of axons compared with the heat-killed iPS-OP group. Values in the graphs represent mean ± SEM. * denote statistical significance compared to heat-killed iPS-OPs by Tukey post hoc tests. Ten images encompassing the injury epicenter from 4 rats were analyzed for each treatment group (P

Fig 8. Behavioral assessment 8 weeks following…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in rats.
The average of the Basso, Beattie, and Bresnahan (BBB) Locomotor Rating Scale (BBB SCORE) for left and right limb movement was evaluated for 55 days following spinal cord injury in rats injected with either PBS (blue), 500,000 heat-killed iPS-OP cells (green), live BC1-OP (black), or MR31-OP (red) cells 24 hrs after injury. Values in the graphs represent mean ± SEM of both left and right limbs as there were no significant differences in response between right and left limb scores. Statistical significance is denoted by a colored asterisk for each cell line with matched color compared to the PBS control group determined by one-way ANOVA followed by a two-way Student’s t-test assuming equal variances (P
All figures (8)
Similar articles
Cited by
References
    1. Gunaseeli I, Doss MX, Antzelevitch C, Hescheler J, Sachinidis A (2010) Induced pluripotent stem cells as a model for accelerated patient- and disease-specific drug discovery. Curr Med Chem 17: 759–766. - PMC - PubMed
    1. Lau F, Ahfeldt T, Osafune K, Akustsu H, Cowan CA (2009) Induced pluripotent stem (iPS) cells: an up-to-the-minute review. F1000 Biol Rep 1: 84 10.3410/B1-84 - DOI - PMC - PubMed
    1. Nishikawa S, Goldstein RA, Nierras CR (2008) The promise of human induced pluripotent stem cells for research and therapy. Nat Rev Mol Cell Biol 9: 725–729. 10.1038/nrm2466 - DOI - PubMed
    1. Wang S, Bates J, Li X, Schanz S, Chandler-Militello D, et al. (2013) Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 12: 252–264. 10.1016/j.stem.2012.12.002 - DOI - PMC - PubMed
    1. Yoshihara H, Shumsky JS, Neuhuber B, Otsuka T, Fischer I, et al. (2006) Combining motor training with transplantation of rat bone marrow stromal cells does not improve repair or recovery in rats with thoracic contusion injuries. Brain Research 1119: 65–75. - PubMed
Show all 78 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig 6. Hematoxylin and eosin and luxol…
Fig 6. Hematoxylin and eosin and luxol fast blue stained paraffin sections of spinal cords from animals subjected to moderate contusive SCI and treated with (A) Heat-killed iPS-OPs or (B) Live, intact iPS-OPs derived from the BC1 line.
As expected for a 12.5 mm contusive injury, cavity formation was seen in both groups; however, the cavitation in spinal cords treated with iPS-OPs did not form well defined glial-like scar structures around the cavity periphery, compared to the control group. Thus, controls showed more pronounced cavitation, which extended further from the epicenter along the central canal. This indicates that the transplantation of live iPS-OPs into the newly formed cavity 24 hours after injury limited the normal expected progression of this level of contusive injury. The asterisks indicate the center of the injury site. Yellow boxes in A and B correspond to adjacent tissue sections stained in C and D, respectively. In these areas immediately surrounding the center of injury, staining with Luxol Fast Blue (LFB) to identify myelin showed reduced staining in the control group (C) versus the iPS-OP transplanted spinal cords (D). Thus, higher levels of LFB in the iPS-OP treated rats may indicate the presence of greater amounts of myelin. The representative spinal cords shown in (A-D) were harvested approximately 2 months after transplantation. Dotted lines help delineate regions of white matter (W) and grey matter (G). (E) Area measurements across groups revealed nearly a 5-fold significant reduction in cavity size in the iPS-OP treated groups compared to the heat-killed controls. Optical density of LBF, normalized to the optical density of the sham control group, was calculated for the grey matter (F) and white matter (G) of spinal sections to quantify the amount of myelin present in each area of the cord. Values in the graphs represent mean ± SEM. * denote statistical significance compared to heat-killed iPS-OPs by Tukey post hoc tests. For each stain, 5 identically numbered tissue slices encompassing the injury epicenter from 4 rats were analyzed for each treatment group, P

Fig 7. Transmission electron microscopy of transverse…

Fig 7. Transmission electron microscopy of transverse sections of the spinal cord showed remyelination of…

Fig 7. Transmission electron microscopy of transverse sections of the spinal cord showed remyelination of spared axons by BC1-derived iPS-OP cells after contusive SCI in rats.
(A) Normal, endogenous myelination of an uninjured rat spinal cord (laminectomy sham control) as indicated by white arrows, showing thick and tightly-packed myelin sheaths. (B) Remyelination observed in rats transplanted with BC1-derived OPs, approximately two months after transplantation. These are sites where oligodendrocytes are detected, as shown in Fig. 4. The relatively thinner and loosely packed myelin sheaths may be compared with the normal condition in (A), suggesting that this is remyelination by human OPs. (C) Multiple nude axons are visible in rats injected with heat-killed iPS-OPs, with no observable remyelination, approximately two months after transplant. (D) Quantification shows that transplantation of BC1-derived OPs resulted in a significant increase in the number of remyelinated axons and reduced demyelinated of axons compared with the heat-killed iPS-OP group. Values in the graphs represent mean ± SEM. * denote statistical significance compared to heat-killed iPS-OPs by Tukey post hoc tests. Ten images encompassing the injury epicenter from 4 rats were analyzed for each treatment group (P

Fig 8. Behavioral assessment 8 weeks following…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in rats.
The average of the Basso, Beattie, and Bresnahan (BBB) Locomotor Rating Scale (BBB SCORE) for left and right limb movement was evaluated for 55 days following spinal cord injury in rats injected with either PBS (blue), 500,000 heat-killed iPS-OP cells (green), live BC1-OP (black), or MR31-OP (red) cells 24 hrs after injury. Values in the graphs represent mean ± SEM of both left and right limbs as there were no significant differences in response between right and left limb scores. Statistical significance is denoted by a colored asterisk for each cell line with matched color compared to the PBS control group determined by one-way ANOVA followed by a two-way Student’s t-test assuming equal variances (P
All figures (8)
Similar articles
Cited by
References
    1. Gunaseeli I, Doss MX, Antzelevitch C, Hescheler J, Sachinidis A (2010) Induced pluripotent stem cells as a model for accelerated patient- and disease-specific drug discovery. Curr Med Chem 17: 759–766. - PMC - PubMed
    1. Lau F, Ahfeldt T, Osafune K, Akustsu H, Cowan CA (2009) Induced pluripotent stem (iPS) cells: an up-to-the-minute review. F1000 Biol Rep 1: 84 10.3410/B1-84 - DOI - PMC - PubMed
    1. Nishikawa S, Goldstein RA, Nierras CR (2008) The promise of human induced pluripotent stem cells for research and therapy. Nat Rev Mol Cell Biol 9: 725–729. 10.1038/nrm2466 - DOI - PubMed
    1. Wang S, Bates J, Li X, Schanz S, Chandler-Militello D, et al. (2013) Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 12: 252–264. 10.1016/j.stem.2012.12.002 - DOI - PMC - PubMed
    1. Yoshihara H, Shumsky JS, Neuhuber B, Otsuka T, Fischer I, et al. (2006) Combining motor training with transplantation of rat bone marrow stromal cells does not improve repair or recovery in rats with thoracic contusion injuries. Brain Research 1119: 65–75. - PubMed
Show all 78 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig 7. Transmission electron microscopy of transverse…
Fig 7. Transmission electron microscopy of transverse sections of the spinal cord showed remyelination of spared axons by BC1-derived iPS-OP cells after contusive SCI in rats.
(A) Normal, endogenous myelination of an uninjured rat spinal cord (laminectomy sham control) as indicated by white arrows, showing thick and tightly-packed myelin sheaths. (B) Remyelination observed in rats transplanted with BC1-derived OPs, approximately two months after transplantation. These are sites where oligodendrocytes are detected, as shown in Fig. 4. The relatively thinner and loosely packed myelin sheaths may be compared with the normal condition in (A), suggesting that this is remyelination by human OPs. (C) Multiple nude axons are visible in rats injected with heat-killed iPS-OPs, with no observable remyelination, approximately two months after transplant. (D) Quantification shows that transplantation of BC1-derived OPs resulted in a significant increase in the number of remyelinated axons and reduced demyelinated of axons compared with the heat-killed iPS-OP group. Values in the graphs represent mean ± SEM. * denote statistical significance compared to heat-killed iPS-OPs by Tukey post hoc tests. Ten images encompassing the injury epicenter from 4 rats were analyzed for each treatment group (P

Fig 8. Behavioral assessment 8 weeks following…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in…

Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in rats.
The average of the Basso, Beattie, and Bresnahan (BBB) Locomotor Rating Scale (BBB SCORE) for left and right limb movement was evaluated for 55 days following spinal cord injury in rats injected with either PBS (blue), 500,000 heat-killed iPS-OP cells (green), live BC1-OP (black), or MR31-OP (red) cells 24 hrs after injury. Values in the graphs represent mean ± SEM of both left and right limbs as there were no significant differences in response between right and left limb scores. Statistical significance is denoted by a colored asterisk for each cell line with matched color compared to the PBS control group determined by one-way ANOVA followed by a two-way Student’s t-test assuming equal variances (P
All figures (8)
Similar articles
Cited by
References
    1. Gunaseeli I, Doss MX, Antzelevitch C, Hescheler J, Sachinidis A (2010) Induced pluripotent stem cells as a model for accelerated patient- and disease-specific drug discovery. Curr Med Chem 17: 759–766. - PMC - PubMed
    1. Lau F, Ahfeldt T, Osafune K, Akustsu H, Cowan CA (2009) Induced pluripotent stem (iPS) cells: an up-to-the-minute review. F1000 Biol Rep 1: 84 10.3410/B1-84 - DOI - PMC - PubMed
    1. Nishikawa S, Goldstein RA, Nierras CR (2008) The promise of human induced pluripotent stem cells for research and therapy. Nat Rev Mol Cell Biol 9: 725–729. 10.1038/nrm2466 - DOI - PubMed
    1. Wang S, Bates J, Li X, Schanz S, Chandler-Militello D, et al. (2013) Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 12: 252–264. 10.1016/j.stem.2012.12.002 - DOI - PMC - PubMed
    1. Yoshihara H, Shumsky JS, Neuhuber B, Otsuka T, Fischer I, et al. (2006) Combining motor training with transplantation of rat bone marrow stromal cells does not improve repair or recovery in rats with thoracic contusion injuries. Brain Research 1119: 65–75. - PubMed
Show all 78 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig 8. Behavioral assessment 8 weeks following…
Fig 8. Behavioral assessment 8 weeks following moderate contusive injury (12.5mm) and cell transplantation in rats.
The average of the Basso, Beattie, and Bresnahan (BBB) Locomotor Rating Scale (BBB SCORE) for left and right limb movement was evaluated for 55 days following spinal cord injury in rats injected with either PBS (blue), 500,000 heat-killed iPS-OP cells (green), live BC1-OP (black), or MR31-OP (red) cells 24 hrs after injury. Values in the graphs represent mean ± SEM of both left and right limbs as there were no significant differences in response between right and left limb scores. Statistical significance is denoted by a colored asterisk for each cell line with matched color compared to the PBS control group determined by one-way ANOVA followed by a two-way Student’s t-test assuming equal variances (P
All figures (8)

References

    1. Gunaseeli I, Doss MX, Antzelevitch C, Hescheler J, Sachinidis A (2010) Induced pluripotent stem cells as a model for accelerated patient- and disease-specific drug discovery. Curr Med Chem 17: 759–766.
    1. Lau F, Ahfeldt T, Osafune K, Akustsu H, Cowan CA (2009) Induced pluripotent stem (iPS) cells: an up-to-the-minute review. F1000 Biol Rep 1: 84 10.3410/B1-84
    1. Nishikawa S, Goldstein RA, Nierras CR (2008) The promise of human induced pluripotent stem cells for research and therapy. Nat Rev Mol Cell Biol 9: 725–729. 10.1038/nrm2466
    1. Wang S, Bates J, Li X, Schanz S, Chandler-Militello D, et al. (2013) Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 12: 252–264. 10.1016/j.stem.2012.12.002
    1. Yoshihara H, Shumsky JS, Neuhuber B, Otsuka T, Fischer I, et al. (2006) Combining motor training with transplantation of rat bone marrow stromal cells does not improve repair or recovery in rats with thoracic contusion injuries. Brain Research 1119: 65–75.
    1. Torres-Espin A, Redondo-Castro E, Hernandez J, Navarro X (2014) Bone marrow mesenchymal stromal cells and olfactory ensheathing cells transplantation after spinal cord injury—a morphological and functional comparison in rats. Eur J Neurosci 39: 1704–1717. 10.1111/ejn.12542
    1. Kumagai G, Tsoulfas P, Toh S, McNiece I, Bramlett HM, et al. (2013) Genetically modified mesenchymal stem cells (MSCs) promote axonal regeneration and prevent hypersensitivity after spinal cord injury. Exp Neurol 248: 369–380. 10.1016/j.expneurol.2013.06.028
    1. Wang LJ, Zhang RP, Li JD (2014) Transplantation of neurotrophin-3-expressing bone mesenchymal stem cells improves recovery in a rat model of spinal cord injury. Acta Neurochir (Wien) 156: 1409–1418. 10.1007/s00701-014-2089-6
    1. Mohammad-Gharibani P, Tiraihi T, Delshad A, Arabkheradmand J, Taheri T (2013) Improvement of contusive spinal cord injury in rats by co-transplantation of gamma-aminobutyric acid-ergic cells and bone marrow stromal cells. Cytotherapy 15: 1073–1085. 10.1016/j.jcyt.2013.05.002
    1. Penha EM, Meira CS, Guimaraes ET, Mendonca MV, Gravely FA, et al. (2014) Use of autologous mesenchymal stem cells derived from bone marrow for the treatment of naturally injured spinal cord in dogs. Stem Cells Int 2014: 437–445.
    1. Reeves A, Keirstead HS (2012) Stem cell based strategies for spinal cord injury repair. Adv Exp Med Biol 760: 16–24.
    1. Quertainmont R, Cantinieaux D, Botman O, Sid S, Schoenen J, et al. (2012) Mesenchymal stem cell graft improves recovery after spinal cord injury in adult rats through neurotrophic and pro-angiogenic actions. PLoS One 7: e39500 10.1371/journal.pone.0039500
    1. Lu P, Kadoya K, Tuszynski MH (2014) Axonal growth and connectivity from neural stem cell grafts in models of spinal cord injury. Curr Opin Neurobiol 27: 103–109. 10.1016/j.conb.2014.03.010
    1. Chang YW, Goff LA, Li H, Kane-Goldsmith N, Tzatzalos E, et al. (2009) Rapid induction of genes associated with tissue protection and neural development in contused adult spinal cord after radial glial cell transplantation. J Neurotrauma 26: 979–993. 10.1089/neu.2008-0762
    1. Ourednik J, Ourednik V, Lynch WP, Schachner M, Snyder EY (2002) Neural stem cells display an inherent mechanism for rescuing dysfunctional neurons. Nat Biotechnol 20: 1103–1110.
    1. Teng YD, Lavik EB, Qu X, Park KI, Ourednik J, et al. (2002) Functional recovery following traumatic spinal cord injury mediated by a unique polymer scaffold seeded with neural stem cells. Proc Natl Acad Sci U S A 99: 3024–3029.
    1. Kerr DA, Llado J, Shamblott MJ, Maragakis NJ, Irani DN, et al. (2003) Human embryonic germ cell derivatives facilitate motor recovery of rats with diffuse motor neuron injury. J Neurosci 23: 5131–5140.
    1. Park DY, Mayle RE, Smith RL, Corcoran-Schwartz I, Kharazi AI, et al. (2013) Combined Transplantation of Human Neuronal and Mesenchymal Stem Cells following Spinal Cord Injury. Global Spine J 3: 1–6. 10.1055/s-0033-1337118
    1. All AH, Bazley FA, Gupta S, Pashai N, Hu C, et al. (2012) Human embryonic stem cell-derived oligodendrocyte progenitors aid in functional recovery of sensory pathways following contusive spinal cord injury. PLoS One 7: e47645 10.1371/journal.pone.0047645
    1. Shah SN, Kerr C, Cope L, Zambidis E, Liu C, et al. (2012) HMGA1 reprograms somatic cells into pluripotent stem cells by inducing stem cell transcriptional networks. PLoS One 7: e48533 10.1371/journal.pone.0048533
    1. Ye Z, Zhan H, Mali P, Dowey S, Williams DM, et al. (2009) Human-induced pluripotent stem cells from blood cells of healthy donors and patients with acquired blood disorders. Blood 114: 5473–5480. 10.1182/blood-2009-04-217406
    1. Chou BK, Mali P, Huang X, Ye Z, Dowey SN, et al. (2011) Efficient human iPS cell derivation by a non-integrating plasmid from blood cells with unique epigenetic and gene expression signatures. Cell Res 21: 518–529. 10.1038/cr.2011.12
    1. Cheng L, Hansen NF, Zhao L, Du Y, Zou C, et al. (2012) Low incidence of DNA sequence variation in human induced pluripotent stem cells generated by nonintegrating plasmid expression. Cell Stem Cell 10: 337–344. 10.1016/j.stem.2012.01.005
    1. Kerr CL, Letzen BS, Hill CM, Agrawal G, Thakor NV, et al. (2010) Efficient differentiation of human embryonic stem cells into oligodendrocyte progenitors for application in a rat contusion model of spinal cord injury. Int J Neurosci 120: 305–313. 10.3109/00207450903585290
    1. Letzen BS, Liu C, Thakor NV, Gearhart JD, All AH, et al. (2010) MicroRNA expression profiling of oligodendrocyte differentiation from human embryonic stem cells. PLoS One 5: e10480 10.1371/journal.pone.0010480
    1. Chaerkady R, Kerr CL, Marimuthu A, Kelkar DS, Kashyap MK, et al. (2009) Temporal analysis of neural differentiation using quantitative proteomics. J Proteome Res 8: 1315–1326. 10.1021/pr8006667
    1. Chaerkady R, Letzen B, Renuse S, Sahasrabuddhe NA, Kumar P, et al. (2011) Quantitative temporal proteomic analysis of human embryonic stem cell differentiation into oligodendrocyte progenitor cells. Proteomics 11: 4007–4020. 10.1002/pmic.201100107
    1. Coetzee T, Fujita N, Dupree J, Shi R, Blight A, et al. (1996) Myelination in the absence of galactocerebroside and sulfatide: normal structure with abnormal function and regional instability. Cell 86: 209–219.
    1. Jakovcevski I, Filipovic R, Mo Z, Rakic S, Zecevic N (2009) Oligodendrocyte development and the onset of myelination in the human fetal brain. Front Neuroanat 3: 1–15. 10.3389/neuro.05.001.2009
    1. Onifer SM, Rabchevsky AG, Scheff SW (2007) Rat models of traumatic spinal cord injury to assess motor recovery. ILAR J 48: 385–395.
    1. Metz GA, Curt A, van de Meent H, Klusman I, Schwab ME, et al. (2000) Validation of the weight-drop contusion model in rats: a comparative study of human spinal cord injury. J Neurotrauma 17: 1–17.
    1. Young W (2002) Spinal cord contusion models. Prog Brain Res 137: 231–255.
    1. Agrawal G, Kerr C, Thakor NV, All AH (2010) Characterization of graded multicenter animal spinal cord injury study contusion spinal cord injury using somatosensory-evoked potentials. Spine (Phila Pa 1976) 35: 1122–1127.
    1. Bazley FA, Hu C, Maybhate A, Pourmorteza A, Pashai N, et al. (2012) Electrophysiological evaluation of sensory and motor pathways after incomplete unilateral spinal cord contusion. J Neurosurg Spine 16: 414–423. 10.3171/2012.1.SPINE11684
    1. Bazley FA, Maybhate A, Tan CS, Thakor NV, Kerr C, et al. (2014) Enhancement of bilateral cortical somatosensory evoked potentials to intact forelimb stimulation following thoracic contusion spinal cord injury in rats. IEEE Trans Neural Syst Rehabil Eng 22: 953–964. 10.1109/TNSRE.2014.2319313
    1. Bazley FA, Pashai N, Kerr CL, All AH (2014) The effects of local and general hypothermia on temperature profiles of the central nervous system following spinal cord injury in rats. Ther Hypothermia Temp Manag 4: 115–124. 10.1089/ther.2014.0002
    1. Maybhate A, Hu C, Bazley FA, Yu Q, Thakor NV, et al. (2012) Potential long-term benefits of acute hypothermia after spinal cord injury: assessments with somatosensory-evoked potentials. Crit Care Med 40: 573–579. 10.1097/CCM.0b013e318232d97e
    1. Basso DM, Beattie MS, Bresnahan JC (1995) A sensitive and reliable locomotor rating scale for open field testing in rats. J Neurotrauma 12: 1–21.
    1. Basso DM, Beattie MS, Bresnahan JC (1996) Graded histological and locomotor outcomes after spinal cord contusion using the NYU weight-drop device versus transection. Exp Neurol 139: 244–256.
    1. Hameed A, Hruban RH, Gage W, Pettis G, Fox WM 3rd (1994) Immunohistochemical expression of CD68 antigen in human peripheral blood T cells. Hum Pathol 25: 872–876.
    1. Tysseling-Mattiace VM, Sahni V, Niece KL, Birch D, Czeisler C, et al. (2008) Self-assembling nanofibers inhibit glial scar formation and promote axon elongation after spinal cord injury. J Neurosci 28: 3814–3823. 10.1523/JNEUROSCI.0143-08.2008
    1. Blight AR (1983) Cellular morphology of chronic spinal cord injury in the cat: analysis of myelinated axons by line-sampling. Neuroscience 10: 521–543.
    1. Durongphongtorn S, McDonell WN, Kerr CL, Neto FJ, Mirakhur KK (2006) Comparison of hemodynamic, clinicopathologic, and gastrointestinal motility effects and recovery characteristics of anesthesia with isoflurane and halothane in horses undergoing arthroscopic surgery. Am J Vet Res 67: 32–42.
    1. Graham V, Khudyakov J, Ellis P, Pevny L (2003) SOX2 functions to maintain neural progenitor identity. Neuron 39: 749–765.
    1. Dawson MR, Levine JM, Reynolds R (2000) NG2-expressing cells in the central nervous system: are they oligodendroglial progenitors? J Neurosci Res 61: 471–479.
    1. Montague P, McCallion AS, Davies RW, Griffiths IR (2006) Myelin-associated oligodendrocytic basic protein: a family of abundant CNS myelin proteins in search of a function. Dev Neurosci 28: 479–487.
    1. Trapp BD, Andrews SB, Cootauco C, Quarles R (1989) The myelin-associated glycoprotein is enriched in multivesicular bodies and periaxonal membranes of actively myelinating oligodendrocytes. J Cell Biol 109: 2417–2426.
    1. Watanabe M, Sakurai Y, Ichinose T, Aikawa Y, Kotani M, et al. (2006) Monoclonal antibody Rip specifically recognizes 2',3'-cyclic nucleotide 3'-phosphodiesterase in oligodendrocytes. J Neurosci Res 84: 525–533.
    1. Parr AM, Kulbatski I, Tator CH (2007) Transplantation of adult rat spinal cord stem/progenitor cells for spinal cord injury. J Neurotrauma 24: 835–845.
    1. Pawelczyk E, Jordan EK, Balakumaran A, Chaudhry A, Gormley N, et al. (2009) In vivo transfer of intracellular labels from locally implanted bone marrow stromal cells to resident tissue macrophages. PLoS One 4: e6712 10.1371/journal.pone.0006712
    1. Fleming JC, Norenberg MD, Ramsay DA, Dekaban GA, Marcillo AE, et al. (2006) The cellular inflammatory response in human spinal cords after injury. Brain 129: 3249–3269.
    1. Lu HZ, Wang YX, Zhou JS, Wang FC, Hu JG (2010) Cyclosporin A increases recovery after spinal cord injury but does not improve myelination by oligodendrocyte progenitor cell transplantation. BMC Neurosci 11: 127 10.1186/1471-2202-11-127
    1. Erceg S, Ronaghi M, Oria M, Rosello MG, Arago MA, et al. (2010) Transplanted oligodendrocytes and motoneuron progenitors generated from human embryonic stem cells promote locomotor recovery after spinal cord transection. Stem Cells 28: 1541–1549. 10.1002/stem.489
    1. George R, Griffin JW (1994) Delayed macrophage responses and myelin clearance during Wallerian degeneration in the central nervous system: the dorsal radiculotomy model. Exp Neurol 129: 225–236.
    1. English K, Mahon BP (2011) Allogeneic mesenchymal stem cells: agents of immune modulation. J Cell Biochem 112: 1963–1968. 10.1002/jcb.23119
    1. Bilic J, Izpisua Belmonte JC (2012) Concise review: Induced pluripotent stem cells versus embryonic stem cells: close enough or yet too far apart? Stem Cells 30: 33–41. 10.1002/stem.700
    1. Hu BY, Weick JP, Yu J, Ma LX, Zhang XQ, et al. (2010) Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc Natl Acad Sci U S A 107: 4335–4340. 10.1073/pnas.0910012107
    1. Doi A, Park IH, Wen B, Murakami P, Aryee MJ, et al. (2009) Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet 41: 1350–1353. 10.1038/ng.471
    1. Gore A, Li Z, Fung HL, Young JE, Agarwal S, et al. (2011) Somatic coding mutations in human induced pluripotent stem cells. Nature 471: 63–67. 10.1038/nature09805
    1. Ronaghi M, Erceg S, Moreno-Manzano V, Stojkovic M (2010) Challenges of stem cell therapy for spinal cord injury: human embryonic stem cells, endogenous neural stem cells, or induced pluripotent stem cells? Stem Cells 28: 93–99. 10.1002/stem.253
    1. Antonic A, Sena ES, Lees JS, Wills TE, Skeers P, et al. (2013) Stem cell transplantation in traumatic spinal cord injury: a systematic review and meta-analysis of animal studies. PLoS Biol 11: e1001738 10.1371/journal.pbio.1001738
    1. Li J, Lepski G (2013) Cell transplantation for spinal cord injury: a systematic review. Biomed Res Int 2013: 786475 10.1155/2013/786475
    1. Krsulovic J, Couve E, Roncagliolo M (1999) Dysmyelination, demyelination and reactive astrogliosis in the optic nerve of the taiep rat. Biol Res 32: 253–262.
    1. Lucchinetti CF, Bruck W, Rodriguez M, Lassmann H (1996) Distinct patterns of multiple sclerosis pathology indicates heterogeneity on pathogenesis. Brain Pathol 6: 259–274.
    1. Wu E, Raine CS (1992) Multiple sclerosis. Interactions between oligodendrocytes and hypertrophic astrocytes and their occurrence in other, nondemyelinating conditions. Lab Invest 67: 88–99.
    1. Keirstead HS, Nistor G, Bernal G, Totoiu M, Cloutier F, et al. (2005) Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J Neurosci 25: 4694–4705.
    1. Urdzikova LM, Ruzicka J, LaBagnara M, Karova K, Kubinova S, et al. (2014) Human mesenchymal stem cells modulate inflammatory cytokines after spinal cord injury in rat. Int J Mol Sci 15: 11275–11293. 10.3390/ijms150711275
    1. Yuan YM, He C (2013) The glial scar in spinal cord injury and repair. Neurosci Bull 29: 421–435. 10.1007/s12264-013-1358-3
    1. Nistor GI, Totoiu MO, Haque N, Carpenter MK, Keirstead HS (2005) Human embryonic stem cells differentiate into oligodendrocytes in high purity and myelinate after spinal cord transplantation. Glia 49: 385–396.
    1. Dasari VR, Spomar DG, Gondi CS, Sloffer CA, Saving KL, et al. (2007) Axonal remyelination by cord blood stem cells after spinal cord injury. J Neurotrauma 24: 391–410.
    1. Brustle O, Jones KN, Learish RD, Karram K, Choudhary K, et al. (1999) Embryonic stem cell-derived glial precursors: a source of myelinating transplants. Science 285: 754–756.
    1. Keirstead HS, Ben-Hur T, Rogister B, O'Leary MT, Dubois-Dalcq M, et al. (1999) Polysialylated neural cell adhesion molecule-positive CNS precursors generate both oligodendrocytes and Schwann cells to remyelinate the CNS after transplantation. J Neurosci 19: 7529–7536.
    1. Czepiel M, Balasubramaniyan V, Schaafsma W, Stancic M, Mikkers H, et al. (2011) Differentiation of induced pluripotent stem cells into functional oligodendrocytes. Glia 59: 882–892. 10.1002/glia.21159
    1. Tokumoto Y, Ogawa S, Nagamune T, Miyake J (2010) Comparison of efficiency of terminal differentiation of oligodendrocytes from induced pluripotent stem cells versus embryonic stem cells in vitro. J Biosci Bioeng 109: 622–628. 10.1016/j.jbiosc.2009.11.013
    1. Tsuji O, Miura K, Okada Y, Fujiyoshi K, Mukaino M, et al. (2010) Therapeutic potential of appropriately evaluated safe-induced pluripotent stem cells for spinal cord injury. Proc Natl Acad Sci U S A 107: 12704–12709. 10.1073/pnas.0910106107
    1. Jang J, Kang HC, Kim HS, Kim JY, Huh YJ, et al. (2011) Induced pluripotent stem cell models from X-linked adrenoleukodystrophy patients. Ann Neurol 70: 402–409. 10.1002/ana.22486
    1. Pouya A, Satarian L, Kiani S, Javan M, Baharvand H (2011) Human induced pluripotent stem cells differentiation into oligodendrocyte progenitors and transplantation in a rat model of optic chiasm demyelination. PLoS One 6: e27925 10.1371/journal.pone.0027925
    1. Zhao T, Zhang ZN, Rong Z, Xu Y (2011) Immunogenicity of induced pluripotent stem cells. Nature 474: 212–215. 10.1038/nature10135

Source: PubMed

3
Abonnere