Skip to main content
Erschienen in: Journal of Neuro-Oncology 2/2017

Open Access 18.10.2016 | Laboratory Investigation

Identification of two distinct mesenchymal stromal cell populations in human malignant glioma

verfasst von: Andreas Svensson, Tania Ramos-Moreno, Sofia Eberstål, Stefan Scheding, Johan Bengzon

Erschienen in: Journal of Neuro-Oncology | Ausgabe 2/2017

Abstract

Gene profiling has revealed that malignant gliomas can be divided into four distinct molecular subtypes, where tumors with a mesenchymal gene expression are correlated with short survival. The present investigation was undertaken to clarify whether human malignant gliomas contain endogenous mesenchymal stromal cells (MSC), fulfilling consensus criteria defined by The International Society for Cellular Therapy, recruited from the host. We found that MSC-like cells can be isolated from primary human malignant gliomas. Two distinct MSC-like cell populations, differing in their expression of the CD90 surface marker, were discovered after cell sorting. RNA sequencing revealed further genetic differences between these two cell populations and MSC-like cells lacking CD90 produced higher amounts of VEGF and PGE2 compared to cells with the true MSC phenotype, implying that the CD90 MSC-like cells most probably are more active in tumor vascularization and immunosuppression than their CD90+ counterpart. The results highlight the CD90 subpopulation as an important tumor component, however, its functional effects in glioma remains to be resolved. Using the protocols presented here, it will be possible to isolate, characterize and analyze brain tumor-derived MSC-like cells in more detail and to further test their functions in vitro and in in vivo xenograft models of glioma.

Introduction

Mesenchymal stromal cells (MSC) are found in a variety of tissues where they constitute a stem cell pool [1]. They can be mobilized to sites of inflammation and angiogenesis, such as wounded tissue and, importantly, various solid cancers [24]. No single MSC-specific marker exists for in vivo detection of this cell type and it has been shown that the marker expression of MSCs greatly depends on the tissue in which they reside [5, 6]. Human MSCs are defined according to in vitro consensus criteria developed by The International Society for Cellular Therapy (ISCT); in culture, MSC should adhere to plastic, they should express CD73, CD90, CD105 but lack expression of CD11b or CD14, CD19 or CD79α, CD34, CD45 and HLA-DR, and they should be able to differentiate into adipocytes, osteoblasts and chondrocytes [7].
MSC constitute a promising vector system for cell mediated gene therapy [8] as they display inherent tumor-tropic migratory capabilities [9, 10] are found in several adult tissues [1] and are easy to isolate and expand in vitro [11]. However the use of MSCs as a tumor-tropic cellular vector system in cancer including brain tumor gene therapy also raise several concerns as it is not clarified how the cells affect the tumor and its microenvironment [12]. Consequently the presence and role of endogenous in human brain tumors has now gained attention [13]. Isolation of glioma-associated human MSCs was recently performed. Analysis of bulk cultures of primary dissociated tissue from human glioma revealed that glioma-associated MSCs were nontumorigenic stromal cells with phenotypical similarities to bone marrow-derived human MSCs [13]. Genomic sequencing of the glioma-associated MSCs suggested that most MSCs were normal non-transformed cells recruited into the glioma.
The present investigation was undertaken in order to expand on these previous recent findings. For this, a robust protocol for the detection of MSCs using 9 phenotypic markers according to the full ISCT consensus criteria has been developed. Following single cell sorting, we here report the findings of two distinct populations of glioma-derived MSC-like cells that differ in their CD90 expression, their gene expression pattern and their production of pro-angiogenic VEGF and immune-suppressive PGE2.

Materials and methods

Tumor samples

Fourteen primary brain tumor samples were obtained from the neurosurgery department at Skåne University Hospital in Lund, Sweden. Ethical permit H15 642/2008. The tissue was washed in phosphate buffered saline (PBS, Life Technologies, Carlsbad, CA, USA), placed in a Petri dish with StemMACS MSC Expansion Media (Miltenyi Biotec, Bergisch Gladbach, Germany) and minced with a scalpel. Afterwards, cells were washed in PBS and then incubated in Accutase (Sigma-Aldrich, Stockholm, Sweden) at 37 °C for 20 min. They were rotated every 5 min to prevent sedimentation. Finally, cells were passed 2–3 times through an 18 G needle before being filtered through a 100 µm nylon mesh. The single cell suspension was put in cell culture flasks with MSC Expansion Media.

Cell culturing

Tumor-derived cells were grown adherently in MSC Expansion Media supplemented with Antibiotic–Antimycotic solution (AAS, Sigma-Aldrich) in cell culturing flasks at 37 °C and 5 % CO2. Half of the medium was changed 1–3 times per week. Cells at 70–100 % confluency were passaged using Accutase.

Flow cytometry

Culture-derived cells at passage 2–4 were stained with the following monoclonal antibodies; Brilliant Violet 421-CD73 (AD2), Alexa Fluor 700-CD90 (5E10, both from Nordic BioSite, Täby, Sweden), PE-CD105 (266), PE-Cy5-HLA-ABC (G46-2.6), FITC-CD14 (M5E2), FITC-CD19 (HIB19), FITC-CD34 (581), FITC-CD45 (HI30) and FITC-HLA-DR (G46-6, all from BD Biosciences) and isotype matched controls for 30 min at 4 °C. TO-PRO-1 (Life Technologies) was used as viability marker and cells were analyzed and sorted on a FACSAria III cell sorter (BD Biosciences, Heidelberg, Germany).

Cell sorting

Culture-derived cells analyzed on the FACSAria III cell sorter were simultaneously sorted based on surface marker expression. Two populations were sorted from each tumor, one expressing the defined MSC phenotype and one expressing the defined MSC phenotype except CD90. Cells were sorted at passage 2–4 and immediately put back in culture.

“In vitro” differentiation

Sorted tumor-derived stromal cells at passage 8–11 were used for in vitro differentiation assays. Bone marrow-derived (BM) MSCs from healthy donors were used as positive control and the experiments were performed as described elsewhere [14].
For adipocyte differentiation, cells were cultured in StemMACS AdipoDiff Media (Miltenyi Biotec) for 14 days. They were fixed with 4 % formaldehyde solution (APL, Stockholm, Sweden) for 60 min at and then incubated in 60 % isopropanol (VWR International, Stockholm, Sweden) for 5 min before being stained with 0.18 % Oil Red O (Sigma-Aldrich) for 5 min, all at room temperature (RT).
For osteoblast differentiation, cells were cultured in osteoblast induction medium for 21 days. They were incubated in ice-cold 70 % ethanol for 1 h at 4 °C before being stained with 40 mM Alizarin Red solution (Sigma-Aldrich) for 10 min at RT.
For chondrocyte differentiation, cell pellets were grown in chondrogenesis induction medium for 28 days. They were fixed in Stefanini’s fixative [15] and then incubated in 20 % sucrose solution (Merck KGaA, Darmstadt, Germany), both at 4 °C overnight. The pellets were frozen in Tissue-Tek O.C.T. Compound (Sakura Finetek Sweden AB, Göteborg, Sweden). 5 µm thick cryosections were permeabilized and blocked with PBS supplemented with 0.3 % Triton X-100, 0.1 % sodium azide, 0.1 % fish skin gelatin (all from Sigma-Aldrich) and 10 % normal donkey serum (Jackson ImmunoResearch Europe Ltd., Suffolk, United Kingdom) for 45 min at RT. They were stained with polyclonal goat anti-human aggrecan antibody (5 µg/ml, R&D Systems) at 4 °C overnight and then Alexa Fluor 594 donkey anti-goat IgG (5 µg/ml, Life Technologies) for 60 min at RT. Nuclei were visualized by incubation with Hoechst 33342 (8.1 µM, Life Technologies) for 15 min at RT.
Stained adipocytes and osteoblasts were analyzed with a Nikon Diaphot 300 and a Nikon D70 (both from Nikon, Yurakucho, Tokyo, Japan). Chondrocyte pellets were analyzed with an Olympus BX61 and an Olympus DP72 (both from Olympus, Shinjuku, Tokyo, Japan).

RNA-sequencing extraction and gene analysis measurements

Total RNA from GBM-47 (passage number 2); GBM-48 (passage number 2); GBM-47-derived MSC-like CD90 cells (passage number 8); GBM-48-derived MSC-like CD90 cells (passage number 5); GBM-47-derived MSC-like CD90+(passage number 10); GBM-48-derived MSC-like CD90+(passage number 6); U87 primary GBM cell line (passage number 30) and human bone-marrow derived MSC (hBM-MSC, passage number 1, from a 61 years old healthy male donor) was isolated by using RNAesay (Qiagen) with DNase treatment. RNA was extracted according to manufacturer instructions and their concentration was determined spectrophotometrically by Nanodrop-ND 1000 spectophotometer (Nanodrop). RNA quality was analyzed using a Bioanalyzer (Agilent) and samples with a RNA integrity greater than seven were further analyzed. Subsequent RNA amplification and analysis were largely performed as previously reported in [16, 17] except that libraries were prepared using TruSeq Stranded mRNA Kit for NeoPrep from Illumina.

ELISA

Sorted tumor-derived stromal cells at passage 10–13 were analyzed for VEGF and PGE2 production using ELISA. 1 × 105 cells/well were plated in a 24 well plate and grown in 1 ml MSC Expansion Media for 24 h. Supernatants were collected, centrifuged and stored at −80 °C prior to analysis. VEGF levels were determined using the Human VEGF DuoSet kit (R&D Systems). PGE2 levels were determined using the Prostaglandin E2 EIA kit (Cayman Chemical Company, Larodan Fine Chemicals, Malmö, Sweden). Experiments were performed in duplicate on three separate occasions.

Statistical analysis

The RNA sequencing data processing was performed using the analysis pipe line method described in SCAN-B article [16]. Thus, after the first filtering processes, only the mRNAs showing differential expression between different groups are taken for further analysis. To compare the mRNA expression profile between the 2 GBM-derived MSC groups, CD90 and CD90+ cells, simple t test was used and a p value <0.01 was considered significant. The VEGF and PGE2 production analysis was performed using Two-way ANOVA, where p < 0.05 was considered statistically significant. Linear regression analysis was performed correlating the survival time of patients to % total MSC in bulk culture or CD90+ cells in bulk culture or CD90 cells in bulk culture in all possible combinatorial forms. A p value lower than 0.05 was considered to be significant.

Results

Cells with MSC marker expression profile are present in human primary brain tumor cultures

Tumor specimens from 14 different glioma patients grown adherently in vitro and displayed a fibroblastic morphology consistent with MSCs (Fig. 1a–c). All 14 tumor samples were grown as bulk cultures. In all of these cultures, large numbers of spindle shaped cells with a morphology fully compatible with MSCs were observed attached to the plastic surface of the culture flask. Cells in bulk cultures were easily expandable, however, since we aimed for sorting at the lowest passage number possible, bulk cultures were never passaged more than a few times. We then assessed, by flow cytometry, whether cells fulfilling the consensus marker expression profile for MSCs are present in human gliomas.
At passage 2–4, all tumors contained a small subpopulation of cells expressing the full MSC phenotype, as analysed by flow cytometry (Fig. 1d–f). Numerous cells displaying the full MSC consensus marker panel except for CD90 were detected. The fraction of MSC-like cells relative to the total number of cells in culture varied within a wide range (Table 1; Fig. 1d–f). Notable was that in the majority of the tumors, the number of cells displaying the CD90 phenotype was larger than the CD90+ population. Routine pathological diagnosis revealed that the tumor with the noticeably highest amount of MSC-like cells was a gliosarcoma. Another interesting finding was that the low-grade astrocytoma (AC-45) contained notably fewer MSC-like cells than most of the high-grade GBMs, however no correlation was observed between patient survival and the % of MSC-like cells in the tumor (data not shown).
Table 1
Fourteen human brain tumors and BM-MSCs analyzed for MSC marker expression using flow cytometry
Sample
Sex
Age (years)*
Diagnosis
Grade
Passage at analysis
MSC** to total cell ratio in culture (%)
CD90 MSC population in culture (%)
Survival (months)
BM-MSC
5
97.1
AC-45
m
31
Astrocytoma
II
4
0.539
3.73
24
ODG-44
f
49
Oligodendro-glioma
III
2
3.62
35.3
***
GBM-40
m
75
GBM
IV
4
13.6
85.4
9
GBM-43
m
78
GBM
IV
4
1.43
21.3
9
GBM-46
m
56
GBM
IV
3
0.417
4.03
3
GBM-47
m
58
GBM
IV
2
7.19
5.58
16
GBM-48
m
71
GBM
IV
2
29.6
63.5
1
GBM-49
f
69
GBM
IV
3
1.33
13.7
10
GBM-51
m
60
GBM
IV
2
1.68
0.525
27
GBM-52
m
76
GBM
IV
4
4.13
6.33
1
GBM-53
m
62
Gliosarcoma
IV
2
34.3
48.7
17
GBM-54
m
69
GBM
IV
2
11.0
27.4
11
GBM-55
f
62
GBM
IV
4
1.94
0.203
8
GBM-56
m
45
GBM
IV
4
0.0183
2.63
***
*Age at the time of surgery
**Positive for CD73, CD90, CD105, HLA class I; negative for CD14, CD19, CD34, CD45, HLA-DR
***Alive

MSC phenotype-expressing cells isolated from human brain tumors can differentiate into osteoblasts and, to some extent, adipocytes and chondrocytes

Next, we determined if the cells expressing MSC markers had the capacity to differentiate into adipocytes, osteoblasts and chondrocytes. Cells were sorted using FACS according to the consensus criteria defined by ISCT. Two populations from each tumor were isolated, one expressing the defined MSC phenotype and one expressing the defined phenotype except CD90. Sorted cells from only two different tumors were able to proliferate in vitro and were thus analyzed further. Before sorting, cells had been passaged for 2–4 times and after sorting these cells were again passaged for a maximal total passage number of 14.
Possible adipocyte differentiation was only detected in the CD90 population of GBM-47 (Fig. 2a). In the CD90+ population, and in the sorted cells from GBM-48, the cells were clearly changed by the adipogenic differentiation stimulation but no clear lipid vacuole staining was visible. Further on, all sorted cells, even the ones lacking CD90, had the capacity to differentiate into osteoblasts (Fig. 2b). Finally, both the CD90 and CD90+ population from GBM-47 formed chondrocytes (Fig. 2c), as assessed by aggrecan immunoreactivity, whereas the corresponding populations from GBM-48 only showed weak aggrecan expression (data not shown).
We conclude that cells displaying a complete MSC phenotype can be present in human brain tumor specimens. MSC-like cells lacking CD90 isolated from primary human brain tumors could differentiate into osteoblasts, adipocytes and chondrocytes, whereas the corresponding cells expressing CD90 only formed osteoblasts and chondrocytes.

Differential mRNA expression in CD90+ and CD90 MSC-like populations

Differentially expressed gene (DEG) analysis of the transcriptional profile associated with the different cell types here analyzed (GBM, GBM-derived MSC, U87 and hBM-MSC) revealed that there is a different mRNA expression profile between the 2 sorted MSC-like cells, CD90+ and CD90 (Fig. 3a). A t test was subsequently performed to identify differentially expressed genes between CD90 and CD90+ cell types, this resulted in total, 211 genes (135 up regulated and 76 down regulated in CD90+ cell lines). Differentially expressed genes were next subjected to functional annotation analysis using Metacore web-based software (https://​portal.​genego.​com/​) and results show that, amongst the up regulated genes in the CD90+ cell lines, 50 % were enriched for the glutathione metabolism; 25 % were enriched for the cytoskeleton remodeling and 8.3 % for cell adhesion. Amongst the down-regulated genes, 46.7 % were involved in apoptosis and survival; 33.3 % were involved in the immune response and 13.3 % in cell adhesion (Fig. 3b–d). Interestingly, and although cell surface expression of CD90 clearly separates two distinct populations of glioma-derived MSC-like cells, the CD90 mRNA expression profile was not significantly different between these two cell populations and thus, was not taken into account for the different analysis (Fig. 3).

The CD90 MSC-like population produces more VEGF and PGE2 compared to its CD90+ counterpart

The two cell populations that were expandable following cell sorting were analyzed for VEGF and PGE2 production. The results show that the CD90 cells from two GBMs secreted significantly higher levels of VEGF compared to the CD90+ cells from the same tumors (mean 166 and 58.6 pg/ml, respectively, p < 0.01; Fig. 4a).
The CD90 population from GBM-47 also produced considerably more PGE2 compared to the CD90+ population from the same tumor (mean 9830 and 234 pg/ml, respectively, p < 0.01). Similar, the PGE2 production of the CD90 population from GBM-48 produced significantly more PGE2 compared to the CD90+ population from the same tumor (mean 213 and 55.5 pg/ml, respectively, p < 0.05; Fig. 4b).

Discussion

Malignant gliomas with a mesenchymal gene expression profile are associated with an invasive growth pattern and short survival and recurring tumors often display a mesenchymal phenotype [18, 19]. In this study, we analyzed 14 primary human malignant gliomas and conclude that all of them harbor cells expressing surface markers defined by ISCT [7].
By employing a stricter flow cytometric analysis, adhering to the ISCT criteria, and by, for the first time for this particular purpose, sorting cells, we expand on previous reports of MSC-like cells in malignant glioma [13]. We found that malignant gliomas harbor two distinct MSC-like cell populations, differing in their CD90 expression. Although a different tri-mesenchymal differentiation capacity was noticed between CD90+ and CD90 populations, as indicated by adipocyte differentiation only in cells lacking CD90, this data is preliminary and further experiments, optimizing the expansion of MSC clones from a larger number of tumor samples, is warranted in order to confirm if the absence of adipocyte differentiation is a distinctive feature of CD90+ cell population.
In most tumor samples, the CD90 population was larger than the CD90+ population. Both populations were expandable and retained their proliferative capacity in vitro. Unfortunately, these cell populations were difficult to detach from the plastic surface during the first passages and we could only perform complete studies in cells derived from 2 patients. The number of MSC-like cells compared to the total number of cells (MSC to total cell number in culture in Table 1) following 2–4 passages in vitro showed a very wide variation. Yet, the present study was not designed in order to quantify the ratio of MSC-like cells within glioma stroma. Interestingly though, the tumor with the highest proportion of MSC-like cells was a gliosarcoma, a GBM subtype with a clear mesenchymal component [14].
To confirm that the two MSC-like cell populations CD90+ and CD90 constitute two distinct cell populations, we analyzed their gene expression profile. Although we were limited in the number of samples, the mRNA expression profiles clearly support the notion that these are, in fact, two different populations. Intriguingly, although the cell surface expression of CD90 separates these populations, their CD90 mRNA expression profile did not differ, suggesting that cell surface glycoprotein CD90 could be either engulfed from the surface due to a cell reaction to some environmental stimuli, thus becoming undetectable on the surface or, that CD90 cells react to some stimuli and overexpress CD90 as a response to the stimuli. In vitro angiogenic stimuli, for example, generate hM-MSC lacking CD90 expression [20] whilst overexpression of surface cell markers upon inflammatory stimuli has been already reported for neurotrophic receptors [21]. In addition, mechanical stress has been reported to decrease CD90 cell surface glycoprotein in MSC [22]. Since little mechanical stress was performed during culturing, and the number of passages were low when mRNA expression analysis was performed, this hypothesis is quite unlikely. Alternatively, the CD90 mRNA could suffer any type of posttranslational preventing the receptor from being visible on the surface [23, 24]. Future experiments for a further deep characterization of CD90+ and CD90 populations are envisaged.
Likewise, the functional significance of CD90 expression, or lack thereof, on glioma-derived MSC-like cells needs further investigations. Previous studies have suggested that CD90+ pericytes within gliomas promote vascularization and immunosuppression [15]. However, CD90 has also been reported to be a tumor suppressor [25]. The present study might lend some support to the latter, as we found that the MSC-like CD90 cells could be producing elevated levels of VEGF compared to the CD90+ cells from the same tumors. If confirmed, this could indicate that the CD90 population of glioma-derived MSC-like cells might be more actively involved in tumor angiogenesis than the CD90+ subpopulation. In addition to its critical role in tumor angiogenesis, VEGF has been reported to attract MSCs themselves to glioma cells [3]. Furthermore, VEGF has been reported to stimulate MSC proliferation [26], indicating that autocrine mechanisms might function to stimulate MSC recruitment and proliferation within glioma.
The immunosuppressive function of BM-MSCs is well characterized and utilized clinically [27]. Among the key immunoactive factors expressed by MSCs is prostaglandin E2 (PGE2) [28], which has been shown to induce immunosuppression within tumors [15, 29]. Because of technical reasons, we were only able to expand FACS sorted cells from two glioblastoma, which severely restricted their further analysis. However, interestingly, the CD90 MSC-like cells secreted markedly higher amounts of PGE2 compared to the CD90+ cells. PGE2 is a molecule known to be important for BM-MSC-induced immunosuppression [30], suggesting that glioma-derived MSC-like cells, and the CD90 subpopulation in particular, may actively contribute to the glioma immunosuppression.
Even though the source of the glioma-derived MSC-like cells is unknown, the most probable explanation is that the tumor recruits them from normal tissue. Inflammatory factors, such as IL-8 [31], monocyte chemotactic protein-1 [32] and stromal cell-derived factor 1α [33] are known to be expressed in gliomas and have been reported as attractants of MSCs in other cancers [34, 35]. Further on, the extensive angiogenesis within malignant gliomas is believed to play a major part in MSC migration. In addition to VEGF, angiogenic factors such as platelet-derived growth factor-BB [36] and transforming growth factor-β1 [3], known to be involved in glioma angiogenesis, have been shown to mediate MSC recruitment [3, 34]. Moreover, we have previously shown that drug-mediated angiogenesis inhibition markedly decreased the migration of BM-MSCs transplanted into experimental rat gliomas [10].
In conclusion, we show that two distinct MSC-like cell populations, differing in their expression of the CD90 surface marker expression, can be isolated from primary human malignant gliomas. Further analysis revealed important differences between these two populations with regards to mRNA expression patterns and, present results show that CD90 cells produced higher amounts of VEGF and PGE2 compared to cells with the true MSC phenotype, implying that CD90 cells may be more active in tumor vascularization and immunosuppression than their CD90+ counterpart. Taken together, our results highlight the CD90 MSC-like subpopulation as an important tumor component, however, its functional effects in glioma remains to be resolved. Using the protocols presented here, it will be possible to isolate, characterize and analyze brain tumor-derived MSC-like cells in more detail and to further test their functions in vitro and in in vivo xenograft models of glioma.

Acknowledgments

The authors would like to thank Teona Roschupkina and Edward Visse for excellent technical flow cytometry support and Thérése Nilsson, Arshad Jamal, Jan C Brune and Roshanak Ghazanfari for sharing their expertise. We are grateful to Translational and Sequencing Unit at Lund Universityfor their contributions to the RNA sequencing analysis. Likewise, we thank Dr. H. Ahlenius, Dr. I. Artner, MSc Jonas Fritze and MSc Tania Singh for their contribution to the mRNA extraction. This work was supported by a donation from Viveca Jeppsson and grants from the Region Skåne Funds, the ALF grant from the Medical Faculty at Lund University, the Swedish Cancer Society, The Crafoord Foundation and the Gunnar Nilsson Cancer Foundation. The Lund Stem Cell Center was supported by a Center of Excellence grant in Life Sciences from the Swedish Foundation for Strategic Research.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Spaeth E, Klopp A, Dembinski J, Andreeff M, Marini F (2008) Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells. Gene Ther 15(10):730–738. doi:10.1038/gt.2008.39 CrossRefPubMed Spaeth E, Klopp A, Dembinski J, Andreeff M, Marini F (2008) Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells. Gene Ther 15(10):730–738. doi:10.​1038/​gt.​2008.​39 CrossRefPubMed
3.
Zurück zum Zitat Birnbaum T, Roider J, Schankin CJ, Padovan CS, Schichor C, Goldbrunner R, Straube A (2007) Malignant gliomas actively recruit bone marrow stromal cells by secreting angiogenic cytokines. J Neurooncol 83(3):241–247. doi:10.1007/s11060-007-9332-4 CrossRefPubMed Birnbaum T, Roider J, Schankin CJ, Padovan CS, Schichor C, Goldbrunner R, Straube A (2007) Malignant gliomas actively recruit bone marrow stromal cells by secreting angiogenic cytokines. J Neurooncol 83(3):241–247. doi:10.​1007/​s11060-007-9332-4 CrossRefPubMed
5.
Zurück zum Zitat Rolandsson S, Andersson Sjoland A, Brune JC, Li H, Kassem M, Mertens F, Westergren A, Eriksson L, Hansson L, Skog I, Bjermer L, Scheding S, Westergren-Thorsson G (2014) Primary mesenchymal stem cells in human transplanted lungs are CD90/CD105 perivascularly located tissue-resident cells. BMJ Open Respir Res 1(1):e000027. doi:10.1136/bmjresp-2014-000027 CrossRefPubMedPubMedCentral Rolandsson S, Andersson Sjoland A, Brune JC, Li H, Kassem M, Mertens F, Westergren A, Eriksson L, Hansson L, Skog I, Bjermer L, Scheding S, Westergren-Thorsson G (2014) Primary mesenchymal stem cells in human transplanted lungs are CD90/CD105 perivascularly located tissue-resident cells. BMJ Open Respir Res 1(1):e000027. doi:10.​1136/​bmjresp-2014-000027 CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Pinho S, Lacombe J, Hanoun M, Mizoguchi T, Bruns I, Kunisaki Y, Frenette PS (2013) PDGFRalpha and CD51 mark human nestin + sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. J Exp Med 210(7):1351–1367. doi:10.1084/jem.20122252 CrossRefPubMedPubMedCentral Pinho S, Lacombe J, Hanoun M, Mizoguchi T, Bruns I, Kunisaki Y, Frenette PS (2013) PDGFRalpha and CD51 mark human nestin + sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. J Exp Med 210(7):1351–1367. doi:10.​1084/​jem.​20122252 CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. CytoTherapy 8(4):315–317. doi:10.1080/14653240600855905 CrossRefPubMed Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. CytoTherapy 8(4):315–317. doi:10.​1080/​1465324060085590​5 CrossRefPubMed
8.
Zurück zum Zitat Nakamura K, Ito Y, Kawano Y, Kurozumi K, Kobune M, Tsuda H, Bizen A, Honmou O, Niitsu Y, Hamada H (2004) Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther 11(14):1155–1164. doi:10.1038/sj.gt.3302276 CrossRefPubMed Nakamura K, Ito Y, Kawano Y, Kurozumi K, Kobune M, Tsuda H, Bizen A, Honmou O, Niitsu Y, Hamada H (2004) Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther 11(14):1155–1164. doi:10.​1038/​sj.​gt.​3302276 CrossRefPubMed
9.
Zurück zum Zitat Bexell D, Gunnarsson S, Svensson A, Tormin A, Henriques-Oliveira C, Siesjo P, Paul G, Salford LG, Scheding S, Bengzon J (2012) Rat multipotent mesenchymal stromal cells lack long-distance tropism to 3 different rat glioma models. Neurosurgery 70(3):731–739. doi:10.1227/NEU.0b013e318232dedd CrossRefPubMed Bexell D, Gunnarsson S, Svensson A, Tormin A, Henriques-Oliveira C, Siesjo P, Paul G, Salford LG, Scheding S, Bengzon J (2012) Rat multipotent mesenchymal stromal cells lack long-distance tropism to 3 different rat glioma models. Neurosurgery 70(3):731–739. doi:10.​1227/​NEU.​0b013e318232dedd​ CrossRefPubMed
10.
Zurück zum Zitat Bexell D, Gunnarsson S, Tormin A, Darabi A, Gisselsson D, Roybon L, Scheding S, Bengzon J (2009) Bone marrow multipotent mesenchymal stroma cells act as pericyte-like migratory vehicles in experimental gliomas. Mol Ther 17(1):183–190. doi:10.1038/mt.2008.229 CrossRefPubMed Bexell D, Gunnarsson S, Tormin A, Darabi A, Gisselsson D, Roybon L, Scheding S, Bengzon J (2009) Bone marrow multipotent mesenchymal stroma cells act as pericyte-like migratory vehicles in experimental gliomas. Mol Ther 17(1):183–190. doi:10.​1038/​mt.​2008.​229 CrossRefPubMed
13.
Zurück zum Zitat Hossain A, Gumin J, Gao F, Figueroa J, Shinojima N, Takezaki T, Priebe W, Villarreal D, Kang SG, Joyce C, Sulman E, Wang Q, Marini FC, Andreeff M, Colman H, Lang FF (2015) Mesenchymal stem cells isolated from human gliomas increase proliferation and maintain stemness of glioma stem cells through the IL-6/gp130/STAT3 pathway. Stem Cells 33(8):2400–2415. doi:10.1002/stem.2053 CrossRefPubMedPubMedCentral Hossain A, Gumin J, Gao F, Figueroa J, Shinojima N, Takezaki T, Priebe W, Villarreal D, Kang SG, Joyce C, Sulman E, Wang Q, Marini FC, Andreeff M, Colman H, Lang FF (2015) Mesenchymal stem cells isolated from human gliomas increase proliferation and maintain stemness of glioma stem cells through the IL-6/gp130/STAT3 pathway. Stem Cells 33(8):2400–2415. doi:10.​1002/​stem.​2053 CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P (2007) The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol (Berl) 114(2):97–109. doi:10.1007/s00401-007-0243-4 CrossRef Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P (2007) The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol (Berl) 114(2):97–109. doi:10.​1007/​s00401-007-0243-4 CrossRef
15.
16.
Zurück zum Zitat Saal LH, Vallon-Christersson J, Hakkinen J, Hegardt C, Grabau D, Winter C, Brueffer C, Tang MH, Reutersward C, Schulz R, Karlsson A, Ehinger A, Malina J, Manjer J, Malmberg M, Larsson C, Ryden L, Loman N, Borg A (2015) The Sweden Cancerome Analysis Network-Breast (SCAN-B) Initiative: a large-scale multicenter infrastructure towards implementation of breast cancer genomic analyses in the clinical routine. Genome Med 7(1):20. doi:10.1186/s13073-015-0131-9 CrossRefPubMedPubMedCentral Saal LH, Vallon-Christersson J, Hakkinen J, Hegardt C, Grabau D, Winter C, Brueffer C, Tang MH, Reutersward C, Schulz R, Karlsson A, Ehinger A, Malina J, Manjer J, Malmberg M, Larsson C, Ryden L, Loman N, Borg A (2015) The Sweden Cancerome Analysis Network-Breast (SCAN-B) Initiative: a large-scale multicenter infrastructure towards implementation of breast cancer genomic analyses in the clinical routine. Genome Med 7(1):20. doi:10.​1186/​s13073-015-0131-9 CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Hakkinen J, Nordborg N, Mansson O, Vallon-Christersson J (2016) Implementation of an Open Source Software solution for Laboratory Information Management and automated RNAseq data analysis in a large-scale Cancer Genomics initiative using BASE with extension package Reggie. bioRxiv. doi:10.1101/038976 Hakkinen J, Nordborg N, Mansson O, Vallon-Christersson J (2016) Implementation of an Open Source Software solution for Laboratory Information Management and automated RNAseq data analysis in a large-scale Cancer Genomics initiative using BASE with extension package Reggie. bioRxiv. doi:10.​1101/​038976
18.
Zurück zum Zitat Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, Misra A, Nigro JM, Colman H, Soroceanu L, Williams PM, Modrusan Z, Feuerstein BG, Aldape K (2006) Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer cell 9(3):157–173. doi:10.1016/j.ccr.2006.02.019 CrossRefPubMed Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, Misra A, Nigro JM, Colman H, Soroceanu L, Williams PM, Modrusan Z, Feuerstein BG, Aldape K (2006) Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer cell 9(3):157–173. doi:10.​1016/​j.​ccr.​2006.​02.​019 CrossRefPubMed
19.
Zurück zum Zitat Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, Alexe G, Lawrence M, O’Kelly M, Tamayo P, Weir BA, Gabriel S, Winckler W, Gupta S, Jakkula L, Feiler HS, Hodgson JG, James CD, Sarkaria JN, Brennan C, Kahn A, Spellman PT, Wilson RK, Speed TP, Gray JW, Meyerson M, Getz G, Perou CM, Hayes DN (2010) Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17(1):98–110. doi:10.1016/j.ccr.2009.12.020 CrossRefPubMedPubMedCentral Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, Alexe G, Lawrence M, O’Kelly M, Tamayo P, Weir BA, Gabriel S, Winckler W, Gupta S, Jakkula L, Feiler HS, Hodgson JG, James CD, Sarkaria JN, Brennan C, Kahn A, Spellman PT, Wilson RK, Speed TP, Gray JW, Meyerson M, Getz G, Perou CM, Hayes DN (2010) Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17(1):98–110. doi:10.​1016/​j.​ccr.​2009.​12.​020 CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Campioni D, Lanza F, Moretti S, Ferrari L, Cuneo A (2008) Loss of Thy-1 (CD90) antigen expression on mesenchymal stromal cells from hematologic malignancies is induced by in vitro angiogenic stimuli and is associated with peculiar functional and phenotypic characteristics. CytoTherapy 10(1):69–82. doi:10.1080/14653240701762364 CrossRefPubMed Campioni D, Lanza F, Moretti S, Ferrari L, Cuneo A (2008) Loss of Thy-1 (CD90) antigen expression on mesenchymal stromal cells from hematologic malignancies is induced by in vitro angiogenic stimuli and is associated with peculiar functional and phenotypic characteristics. CytoTherapy 10(1):69–82. doi:10.​1080/​1465324070176236​4 CrossRefPubMed
21.
Zurück zum Zitat Becher B, Antel JP (1996) Comparison of phenotypic and functional properties of immediately ex vivo and cultured human adult microglia. Glia 18(1):1–10CrossRefPubMed Becher B, Antel JP (1996) Comparison of phenotypic and functional properties of immediately ex vivo and cultured human adult microglia. Glia 18(1):1–10CrossRefPubMed
24.
Zurück zum Zitat Gui M, Wiest DL, Li J, Kappes D, Hardy RR, Hayakawa K (1999) Peripheral CD4+ T cell maturation recognized by increased expression of Thy-1/CD90 bearing the 6C10 carbohydrate epitope. J Immunol (Baltimore, Md: 1950) 163(9):4796–4804 Gui M, Wiest DL, Li J, Kappes D, Hardy RR, Hayakawa K (1999) Peripheral CD4+ T cell maturation recognized by increased expression of Thy-1/CD90 bearing the 6C10 carbohydrate epitope. J Immunol (Baltimore, Md: 1950) 163(9):4796–4804
25.
Zurück zum Zitat Abeysinghe HR, Cao Q, Xu J, Pollock S, Veyberman Y, Guckert NL, Keng P, Wang N (2003) THY1 expression is associated with tumor suppression of human ovarian cancer. Cancer Genet Cytogenet 143(2):125–132CrossRefPubMed Abeysinghe HR, Cao Q, Xu J, Pollock S, Veyberman Y, Guckert NL, Keng P, Wang N (2003) THY1 expression is associated with tumor suppression of human ovarian cancer. Cancer Genet Cytogenet 143(2):125–132CrossRefPubMed
28.
Zurück zum Zitat Spaggiari GM, Abdelrazik H, Becchetti F, Moretta L (2009) MSCs inhibit monocyte-derived DC maturation and function by selectively interfering with the generation of immature DCs: central role of MSC-derived prostaglandin E2. Blood 113(26):6576–6583. doi:10.1182/blood-2009-02-203943 CrossRefPubMed Spaggiari GM, Abdelrazik H, Becchetti F, Moretta L (2009) MSCs inhibit monocyte-derived DC maturation and function by selectively interfering with the generation of immature DCs: central role of MSC-derived prostaglandin E2. Blood 113(26):6576–6583. doi:10.​1182/​blood-2009-02-203943 CrossRefPubMed
29.
Zurück zum Zitat Djouad F, Plence P, Bony C, Tropel P, Apparailly F, Sany J, Noel D, Jorgensen C (2003) Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood 102(10):3837–3844. doi:10.1182/blood-2003-04-1193 CrossRefPubMed Djouad F, Plence P, Bony C, Tropel P, Apparailly F, Sany J, Noel D, Jorgensen C (2003) Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood 102(10):3837–3844. doi:10.​1182/​blood-2003-04-1193 CrossRefPubMed
30.
31.
Zurück zum Zitat Kim DS, Kim JH, Lee JK, Choi SJ, Kim JS, Jeun SS, Oh W, Yang YS, Chang JW (2009) Overexpression of CXC chemokine receptors is required for the superior glioma-tracking property of umbilical cord blood-derived mesenchymal stem cells. Stem Cells Dev 18(3):511–519. doi:10.1089/scd.2008.0050 CrossRefPubMed Kim DS, Kim JH, Lee JK, Choi SJ, Kim JS, Jeun SS, Oh W, Yang YS, Chang JW (2009) Overexpression of CXC chemokine receptors is required for the superior glioma-tracking property of umbilical cord blood-derived mesenchymal stem cells. Stem Cells Dev 18(3):511–519. doi:10.​1089/​scd.​2008.​0050 CrossRefPubMed
32.
Zurück zum Zitat Dwyer RM, Potter-Beirne SM, Harrington KA, Lowery AJ, Hennessy E, Murphy JM, Barry FP, O’Brien T, Kerin MJ (2007) Monocyte chemotactic protein-1 secreted by primary breast tumors stimulates migration of mesenchymal stem cells. Clin Cancer Res 13(17):5020–5027. doi:10.1158/1078-0432.ccr-07-0731 CrossRefPubMed Dwyer RM, Potter-Beirne SM, Harrington KA, Lowery AJ, Hennessy E, Murphy JM, Barry FP, O’Brien T, Kerin MJ (2007) Monocyte chemotactic protein-1 secreted by primary breast tumors stimulates migration of mesenchymal stem cells. Clin Cancer Res 13(17):5020–5027. doi:10.​1158/​1078-0432.​ccr-07-0731 CrossRefPubMed
33.
Zurück zum Zitat Menon LG, Picinich S, Koneru R, Gao H, Lin SY, Koneru M, Mayer-Kuckuk P, Glod J, Banerjee D (2007) Differential gene expression associated with migration of mesenchymal stem cells to conditioned medium from tumor cells or bone marrow cells. Stem Cells 25(2):520–528. doi:10.1634/stemcells.2006-0257 CrossRefPubMed Menon LG, Picinich S, Koneru R, Gao H, Lin SY, Koneru M, Mayer-Kuckuk P, Glod J, Banerjee D (2007) Differential gene expression associated with migration of mesenchymal stem cells to conditioned medium from tumor cells or bone marrow cells. Stem Cells 25(2):520–528. doi:10.​1634/​stemcells.​2006-0257 CrossRefPubMed
35.
Zurück zum Zitat Xu F, Shi J, Yu B, Ni W, Wu X, Gu Z (2010) Chemokines mediate mesenchymal stem cell migration toward gliomas in vitro. Oncol Rep 23(6):1561–1567PubMed Xu F, Shi J, Yu B, Ni W, Wu X, Gu Z (2010) Chemokines mediate mesenchymal stem cell migration toward gliomas in vitro. Oncol Rep 23(6):1561–1567PubMed
36.
Zurück zum Zitat Nakamizo A, Marini F, Amano T, Khan A, Studeny M, Gumin J, Chen J, Hentschel S, Vecil G, Dembinski J, Andreeff M, Lang FF (2005) Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res 65(8):3307–3318. doi:10.1158/0008-5472.can-04-1874 PubMed Nakamizo A, Marini F, Amano T, Khan A, Studeny M, Gumin J, Chen J, Hentschel S, Vecil G, Dembinski J, Andreeff M, Lang FF (2005) Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res 65(8):3307–3318. doi:10.​1158/​0008-5472.​can-04-1874 PubMed
Metadaten
Titel
Identification of two distinct mesenchymal stromal cell populations in human malignant glioma
verfasst von
Andreas Svensson
Tania Ramos-Moreno
Sofia Eberstål
Stefan Scheding
Johan Bengzon
Publikationsdatum
18.10.2016
Verlag
Springer US
Erschienen in
Journal of Neuro-Oncology / Ausgabe 2/2017
Print ISSN: 0167-594X
Elektronische ISSN: 1573-7373
DOI
https://doi.org/10.1007/s11060-016-2302-y

Weitere Artikel der Ausgabe 2/2017

Journal of Neuro-Oncology 2/2017 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.