Summary

Processing of Primary Brain Tumor Tissue for Stem Cell Assays and Flow Sorting

Published: September 25, 2012
doi:

Summary

The identification of brain tumor initiating cells (BTICs), the rare cells within a heterogeneous tumor possessing stem cell properties, provides new insights into human brain tumor pathogenesis. We have refined specific culture conditions to enrich for BTICs, and we routinely use flow cytometry to further enrich these populations. Self-renewal assays and transcript analysis by single cell RT-PCR can subsequently be performed on these isolated cells.

Abstract

Brain tumors are typically comprised of morphologically diverse cells that express a variety of neural lineage markers. Only a relatively small fraction of cells in the tumor with stem cell properties, termed brain tumor initiating cells (BTICs), possess an ability to differentiate along multiple lineages, self-renew, and initiate tumors in vivo. We applied culture conditions originally used for normal neural stem cells (NSCs) to a variety of human brain tumors and found that this culture method specifically selects for stem-like populations. Serum-free medium (NSC) allows for the maintenance of an undifferentiated stem cell state, and the addition of bFGF and EGF allows for the proliferation of multi-potent, self-renewing, and expandable tumorspheres.

To further characterize each tumor’s BTIC population, we evaluate cell surface markers by flow cytometry. We may also sort populations of interest for more specific characterization. Self-renewal assays are performed on single BTICs sorted into 96 well plates; the formation of tumorspheres following incubation at 37 °C indicates the presence of a stem or progenitor cell. Multiple cell numbers of a particular population can also be sorted in different wells for limiting dilution analysis, to analyze self-renewal capacity. We can also study differential gene expression within a particular cell population by using single cell RT-PCR.

The following protocols describe our procedures for the dissociation and culturing of primary human samples to enrich for BTIC populations, as well as the dissociation of tumorspheres. Also included are protocols for staining for flow cytometry analysis or sorting, self-renewal assays, and single cell RT-PCR.

Introduction

Brain tumors are among the most aggressive and heterogeneous cancers known in humans. Although their earlier detection and diagnosis have been facilitated by modern neuro-imaging technology, we still lack curative therapies for many brain tumors, particularly for diffuse, invasive ones or those located deep in the brain.

Brain tumors represent the leading cause of cancer mortality in children due to their highly aggressive and often incurable nature. Glioblastoma (GBM), the most common primary brain tumor in adults, is one of the most aggressive human cancers, feared for its uniformly fatal prognosis1. This highly malignant astrocytic tumor (WHO Grade 4) usually occurs in the cerebral hemispheres of adults, and can also occur in young children and infants. Its growth is rapid and infiltrative, and diagnostic pathological features include nuclear pleomorphism, microvascular proliferation, and necrosis2,3. For adults with newly diagnosed GBM, median survival rarely extends beyond 12 months1, with generally poor responses to all therapeutic modalities. We noted that there are many functional and genetic similarities shared by somatic stem cells and cancer cells, and that the molecular pathways that regulate normal brain development are often dysregulated in cancer. In applying stem cell biology paradigms to the study of brain tumors, we were the first researchers to prospectively identify and purify a subpopulation of cells from human GBMs which exhibited the stem cell properties of proliferation, self-renewal, and differentiation in vitro4 and in vivo5. We applied culture conditions and assays originally used to characterize normal neural stem cells (NSCs) in vitro 6,7 to multiple pediatric and adult brain tumors, and enriched for these stem-like cells by cell sorting for the neural progenitor cell surface marker CD1338,9. The CD133+ brain tumor fraction contained cells that had a much higher frequency of tumor initiation than the CD133- fraction in NOD-SCID mouse brains5,10. This formally established that only a rare subset of brain tumor cells with stem cell properties are tumor-initiating, earning them the name “brain tumor initiating cells” or “BTICs”. The novel identification of BTICs provides new insights into human brain tumorigenesis, giving strong support for the cancer stem cell hypothesis10-13 as the basis for many solid tumors, and establishes a novel cellular target for more effective cancer therapies14-20. Therapies that focus on killing the bulk of the tumor may miss the rare stem-like fraction, allowing the tumor to continue to grow. Therapies that focus on killing the cancer stem cell may provide better treatment and prognosis for patients with brain tumors.

In order to study BTIC populations, we have refined our culture protocols to specifically select for cell populations within human brain tumors that possess stem cell properties. Serum-free, neural stem cell (NSC) medium allows for the maintenance of an undifferentiated stem cell state, and the addition of basic fibroblast growth factor (bFGF), epidermal growth factor (EGF), and leukemia inhibitory factor (LIF) allows for the proliferation of multi-potent, self-renewing, and expandable human tumorspheres. Here, we describe the methods involved in processing of primary brain tumors and culturing them in NSC medium to enrich for BTIC populations. We have called our experimental model system “BTIC patient isolates” to emphasize the fact that these cells are only minimally cultured under stem cell conditions to select for stem cell populations. Subsequent immunolabelling of BTIC populations for key stem cell markers such as CD133 and CD15 and flow cytometry analysis is also described. We then discuss the limiting dilution analysis, which aids in studying the self-renewal potential of BTICs. Finally, we explore the gene expression analysis of these rare cells by sorting single cells onto AmpliGrid slides and performing single cell RT-PCR. These techniques are also applicable to other brain tumors such as medulloblastoma, ependymoma and pediatric gliomas.

Protocol

1. Culture of Brain Tumor Tissue Add 200 μl thawed Liberase (Roche Applied Science) to 15 ml of artificial CSF (aCSF- see Table 1) and place into 37 °C water bath. Liberase TM is a mix of proteolytic enzymes used to dissociate primary tissue samples, as well as cultured tumorspheres. Unlike Trypsin-EDTA, the Liberase method preserves the surface antigen CD133. For a tissue sample of about 0.5 cm3, we use 200 μl of Liberase. If the tissue is smaller, we use 100 ul. <…

Discussion

The cancer stem cell hypothesis10, based on work in leukemia21, breast cancer11 and brain cancer 4,5, suggests that only a relatively small fraction of cells in the tumor, termed cancer stem cells, possess an ability to extensively proliferate and self-renew. Most of the tumor cells lose the ability to proliferate and self-renew as they differentiate into cells that become the phenotypic signature of the tumor. Finding the key cells in the brain tumor population that are able t…

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was funded by the Ontario Institute of Cancer Research (OICR), the Terry Fox Foundation and the American Association of Neurological Surgeons.

Materials

Name of the reagent Company Catalogue number
1:1 DMEM:F12 Invitrogen 11320-082
N2 supplement Invitrogen 17502-048
1M HEPES Wisent 330-050-EL
Glucose Invitrogen 15023-021
N-acetylcysteine Sigma Aldrich A9165-25g
Neural survival factor -1 (NSF-1) Lonza Clonetics CC-4323
Epidermal growth factor (EGF) Sigma Aldrich E9644
Basic fibroblast growth factor (bFGF) Invitrogen PHG0261
Leukemia inhibitory factor (LIF) Millipore LIF1010
Antibiotic/mycotic Wisent 450-115-EL
Liberase TM Roche 05 401 119 001
Ammonium chloride solution Stem Cell Technologies 07850

References

  1. Ohgaki, H., Kleihues, P. Epidemiology and etiology of gliomas. Acta Neuropathol. 109, 93 (2005).
  2. Huse, J. T., Holland, E. C. Targeting brain cancer: advances in the molecular pathology of malignant glioma. 10, 319 (2010).
  3. Wechsler-Reya, R., Scott, M. P. The developmental biology of brain tumors. Annu. Rev. Neurosci. 24, 385 (2001).
  4. Singh, S. K. Identification of a cancer stem cell in human brain tumors. Cancer Res. 63, 5821 (2003).
  5. Singh, S. K. Identification of human brain tumour initiating cells. Nature. 432, 396 (2004).
  6. Reynolds, B. A., Weiss, S. Clonal and population analyses demonstrate that an EGF-responsive mammalian embryonic CNS precursor is a stem cell. Dev. Biol. 175, 1 (1996).
  7. Tropepe, V. Distinct neural stem cells proliferate in response to EGF and FGF in the developing mouse telencephalon. Dev. Biol. 208, 166 (1999).
  8. Yin, A. H. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood. 90, 5002 (1997).
  9. Yu, Y., Flint, A., Dvorin, E. L., Bischoff, J. AC133-2, a novel isoform of human AC133 stem cell antigen. J. Biol. Chem. 277, 20711 (2002).
  10. Reya, T., Morrison, S. J., Clarke, M. F., Weissman, I. L. Stem cells, cancer, and cancer stem cells. Nature. 414, 105 (2001).
  11. Al-Hajj, M. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. U. S. A. 100, 3983 (2003).
  12. Bonnet, D., Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 3, 730 (1997).
  13. Matsui, W. Characterization of clonogenic multiple myeloma cells. Blood. 103, 2332 (2004).
  14. Bao, S. Targeting cancer stem cells through L1CAM suppresses glioma growth. Cancer Res. 68, 6043 (2008).
  15. Bao, S. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 444, 756 (2006).
  16. Bao, S. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 66, 7843 (2006).
  17. Beier, D. Temozolomide preferentially depletes cancer stem cells in glioblastoma. Cancer Res. 68, 5706 (2008).
  18. Piccirillo, S. G. Distinct pools of cancer stem-like cells coexist within human glioblastomas and display different tumorigenicity and independent genomic evolution. Oncogene. 28, 1807 (2009).
  19. Piccirillo, S. G. morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature. 444, 761 (2006).
  20. Rich, J. N., Bao, S. Chemotherapy and cancer stem cells. Cell Stem Cell. 1, 353 (2007).
  21. Lapidot, T. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 367, 645 (1994).
  22. Reya, T., Morrison, S. J., Clarke, M. F., Weissman, I. L. Stem cells, cancer, and cancer stem cells. Nature. 414, 105 (2001).
  23. Fuchs, E., Segre, J. A. Stem cells: a new lease on life. Cell. 100, 143 (2000).
  24. Weissman, I. L. Stem cells: units of development, units of regeneration, and units in evolution. Cell. 100, 157 (2000).
  25. Reynolds, B. A., Weiss, S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science. 255, 1707 (1992).
  26. Reynolds, B. A., Tetzlaff, W., Weiss, S. A multipotent EGF-responsive striatal embryonic progenitor cell produces neurons and astrocytes. J. Neurosci. 12, 4565 (1992).
  27. Hulett, H. R., Bonner, W. A., Barrett, J., Herzenberg, L. A. Cell sorting: automated separation of mammalian cells as a function of intracellular fluorescence. Science. 166, 747 (1969).
  28. Kohler, G., Milstein, C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 256, 495 (1975).
  29. Barrett, L. E. Self-renewal does not predict tumor growth potential in mouse models of high-grade glioma. Cancer Cell. 21, 11 (2012).
  30. Venugopal, C. Bmi1 marks intermediate precursors during differentiation of human brain tumor initiating cells. Stem Cell Res. 8, 141 (2012).
  31. Gerlinger, M. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883 (2012).
  32. Gilbertson, R. J. Medulloblastoma: signalling a change in treatment. Lancet. Oncol. 5, 209 (2004).
  33. Zhu, Y., Parada, L. F. The molecular and genetic basis of neurological tumours. Nat. Rev. Cancer. 2, 616 (2002).
  34. Maher, E. A. Malignant glioma: genetics and biology of a grave matter. Genes Dev. 15, 1311 (2001).
  35. Blake, W. J., KAErn, M., Cantor, C. R., Collins, J. J. Noise in eukaryotic gene expression. Nature. 422, 633 (2003).
  36. Elowitz, M. B., Levine, A. J., Siggia, E. D., Swain, P. S. Stochastic gene expression in a single cell. Science. 297, 1183 (2002).
  37. Maheshri, N., O’Shea, E. K. Living with noisy genes: how cells function reliably with inherent variability in gene expression. Annu. Rev. Biophys. Biomol. Struct. 36, 413 (2007).
  38. Raj, A. Stochastic mRNA synthesis in mammalian cells. PLoS Biol. 4, e309 (2006).
  39. Ross, I. L., Browne, C. M., Hume, D. A. Transcription of individual genes in eukaryotic cells occurs randomly and infrequently. Immunol. Cell Biol. 72, 177 (1994).
  40. Kubista, M. The real-time polymerase chain reaction. Mol. Aspects Med. 27, 95 (2006).
  41. Nolan, T., Hands, R. E., Bustin, S. A. Quantification of mRNA using real-time RT-PCR. Nat. Protoc. 1, 1559 (2006).
check_url/4111?article_type=t

Play Video

Cite This Article
Venugopal, C., McFarlane, N. M., Nolte, S., Manoranjan, B., Singh, S. K. Processing of Primary Brain Tumor Tissue for Stem Cell Assays and Flow Sorting. J. Vis. Exp. (67), e4111, doi:10.3791/4111 (2012).

View Video