Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Gliomagenesis: genetic alterations and mouse models

Key Points

  • Gliomas are primary tumours that arise from glial cells in the brain and spinal cord. The most malignant gliomas — glioblastoma multiforme (GBM) — are nearly always fatal. Treatment strategies for this disease have remained unchanged for many years and most are based on a limited understanding of the biology of the disease.

  • Chromosomal instability and the deletion and amplification of certain genes are a hallmark of the more severe clinical grades of human glioma.

  • The cells of malignant gliomas share certain characteristics with undifferentiated glial progenitor cells. Mutations found in GBMs frequently activate the signalling pathways that control the differentiation and proliferation of these progenitors or disrupt cell-cycle arrest pathways.

  • Recapitulating the genetic alterations found in human gliomas in mouse models gives rise to tumours that histologically resemble human gliomas. These mouse models have given clues as to the molecular origins of gliomas, and should contribute to the design and testing of new rational therapies.

Abstract

Glioblastoma multiforme is the most malignant of the primary brain tumours and is almost always fatal. The treatment strategies for this disease have not changed appreciably for many years and most are based on a limited understanding of the biology of the disease. However, in the past decade, characteristic genetic alterations have been identified in gliomas that might underlie the initiation or progression of the disease. Recent modelling experiments in mice are helping to delineate the molecular aetiology of this disease and are providing systems to identify and test novel and rational therapeutic strategies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Cellular differentiation in the central nervous system.
Figure 2: Signalling pathways altered by mutations in human gliomas.
Figure 3: INK4A-initiated cell-cycle arrest pathways.
Figure 4: Pathways to gliomagenesis.
Figure 5: Mouse glioma histology.

Similar content being viewed by others

References

  1. Kleihues, P. & Cavenee, W. Pathology and genetics of tumors of the nervous system. (IARC Press, Lyon, 2000). An excellent overview of the molecular and histological characteristics of brain tumours.

  2. Russel, D. S. & Rubenstein, L. J. Pathology of tumors of the nervous system. (Williams & Wilkins, Baltimore, 1989).Classic in-depth text on tumour pathology in the central nervous system.

  3. Scherer, H. J. Structural development in gliomas. Am. J. Cancer 34 , 333–351 (1938).

    Google Scholar 

  4. Jensen, R. Growth factor-mediated angiogenesis in the malignant progression of glial tumors: a review. Surg. Neurol. 49, 189– 195 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Uhm, J. H., Dooley, N. P., Villemure, J. G. & Yong, V. W. Mechanisms of glioma invasion: role of matrix-metalloproteinases. Can. J. Neurol. Sci. 24, 3–15 (1997).

    Article  CAS  PubMed  Google Scholar 

  6. Gage, F. H. Mammalian neural stem cells. Science 287, 1433–1438 (2000). Review of cell lineage specification in the central nervous system during development and in adulthood.

    Article  CAS  PubMed  Google Scholar 

  7. Lee, J. C., Mayer-Proschel, M. & Rao, M. S. Gliogenesis in the central nervous system. Glia 30, 105–121 ( 2000).

    Article  CAS  PubMed  Google Scholar 

  8. Mi, H. & Barres, B. Purification and characterization of astrocyte precursor cells in the developing rat optic nerve. J. Neurosci. 19, 1049–1061 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cameron, H. A., Hazel, T. G. & McKay, R. D. Regulation of neurogenesis by growth factors and transmitters. J. Neurobiol. 36, 287– 309 (1998).

    Article  CAS  PubMed  Google Scholar 

  10. McKinnon, R. D., Matsui, T., Dubois-Dalcq, M. & Aaronson, S. A. FGF modulates the PDGF-driven pathway of oligodendrocyte development. Neuron 5, 603–614 ( 1990).

    Article  CAS  PubMed  Google Scholar 

  11. Mayer, M., Bogler, O. & Nobel, M. The inhibition of oligodendrocytic differentiation of O2A progenitors caused by basic fibroblast growth factor is overridden by astrocytes. Glia 8, 12– 19 (1993).

    Article  CAS  PubMed  Google Scholar 

  12. Bogler, O., Wren, D., Barnett, S., Land, H. & Nobel, M. Cooperation between two growth factors promotes extended self-renewal and inhibits differentiation of oligodendrocyte-type 2 astrocyte (O2A) progenitor cells. Proc. Natl Acad. Sci. USA 87, 6368 –6372 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Mayer, M., Bhakoo, K. & Nobel, M. Ciliary neurotrophic factor and leukemia inhibitory factor promote the generation, maturation and survival of oligodendrocytes in vitro. Development 120, 143– 153 (1994).

    CAS  PubMed  Google Scholar 

  14. Rajan, P. & McKay, R. D. Multiple routes to astrocytic differentiation in the CNS. J. Neurosci. 18, 3620– 3629 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Nobel, M. & Mayer-Proschel, M. Growth factors, glia and gliomas. J. Neurooncol. 35, 193– 209 (1997).Review of the connections between glial cell differentiation and gliomagenesis.

    Article  Google Scholar 

  16. Takahashi, J. et al. Correlation of basic fibroblast growth factor expression levels with the degree of malignancy and vascularity in human gliomas. J. Neurosurg. 76, 792–798 (1992).

    Article  CAS  PubMed  Google Scholar 

  17. Ekstrand, A. J. et al. Genes for epidermal growth factor receptor, transforming growth factor α, and epidermal growth factor and their expression in human gliomas in vivo. Cancer Res. 51, 2164–2172 (1991).

    CAS  PubMed  Google Scholar 

  18. Weis, J. et al. CNTF and its receptor subunits in human gliomas. J. Neurooncol. 44, 243–253 (1999).

    Article  CAS  PubMed  Google Scholar 

  19. Guha, A., Dashner, K., Black, P. M., Wagner, J. A. & Stiles, C. D. Expression of PDGF and PDGF receptors in human astrocytoma operation specimens supports the existence of an autocrine loop. Int. J. Cancer 60, 168– 173 (1995).

    Article  CAS  PubMed  Google Scholar 

  20. Wong, A. J. et al. Structural alterations of the epidermal growth factor receptor gene in human gliomas. Proc. Natl Acad. Sci. USA 89 , 2965–2969 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hunter, T. Protein kinases and phosphatases: the yin and yang of protein phosphorylation and signaling. Cell 80, 225– 236 (1995).

    Article  CAS  PubMed  Google Scholar 

  22. Martin-Blanco, E. p38 MAPK signalling cascades: ancient roles and new functions. Bioessays 22, 637–645 ( 2000).

    Article  CAS  PubMed  Google Scholar 

  23. Xu, G. F. The catalytic domain of the neurofibromatosis type 1 gene product stimulates ras GTPase and complements ira mutants of S. cerevisiae. Cell 63, 835–841 ( 1990).

    Article  CAS  PubMed  Google Scholar 

  24. Guha, A., Feldkamp, M. M., Lau, N., Boss, G. & Pawson, A. Proliferation of human malignant astrocytomas is dependent on Ras activation. Oncogene 15, 2755– 2765 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Jiang, B. H., Aoki, M., Zheng, J. Z., Li, J. & Vogt, P. K. Myogenic signaling of phosphatidylinositol 3-kinase requires the serine-threonine kinase Akt/protein kinase B. Proc. Natl Acad. Sci. USA 96, 2077–2081 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Thomas, G. & Hall, M. N. TOR signaling and control of cell growth. Curr. Opin. Cell Biol. 9, 782– 787 (1997).

    Article  CAS  PubMed  Google Scholar 

  27. Fujisawa, H. et al. Acquisition of the glioblastoma phenotype during astrocytoma progression is associated with loss of heterozygosity on 10q25-qter. Am. J. Pathol. 155, 387–394 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sano, T. et al. Differential expression of MMAC/PTEN in glioblastoma multiforme: relationship to localization and prognosis. Cancer Res. 59, 1820–1824 (1999).

    CAS  PubMed  Google Scholar 

  29. Holland, E. C. et al. Combined activation of Ras and Akt in neural progenitors induces glioblastoma formation in mice. Nature Genet. 12, 55–57 (2000).This study demonstrated the cooperation between the Ras and Akt signalling pathways in the formation of GBMs in a mouse model. PubMed

    Article  Google Scholar 

  30. Ichimura, K., Schmidt, E. E., Goike, H. M. & Collins, V. P. Human glioblastomas with no alterations of the CDKN2A (p16INK4A, MTS1) and CDK4 genes have frequent mutations of the retinoblastoma gene. Oncogene 13, 1065–1072 ( 1996).A large series of gliomas were analysed for mutations in this study, illustrating the importance of cell-cycle arrest pathways in GBM formation.

    CAS  PubMed  Google Scholar 

  31. Jen, J. et al. Deletion of p16 and p15 genes in brain tumors. Cancer Res. 54, 6353–6358 (1994).

    CAS  PubMed  Google Scholar 

  32. Serrano, M., Hannon, G. J. & Beach, D. A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature 366, 704–707 (1993).

    Article  CAS  PubMed  Google Scholar 

  33. Kamijo, T. et al. Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19Arf. Cell 91, 649–659 (1993). PubMed

    Article  Google Scholar 

  34. Pomerantz, J. et al. The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2's inhibition of p53 . Cell 92, 713–723 (1998).

    Article  CAS  PubMed  Google Scholar 

  35. Haber, D. A. Splicing into senescence: the curious case of p16 and p19ARF. Cell 91, 555–558 ( 1997).

    Article  CAS  PubMed  Google Scholar 

  36. Sherr, C. J. & Weber, J. D. The ARF/p53 pathway. Curr. Opin. Genet. Dev. 10, 94–99 (2000).

    Article  CAS  PubMed  Google Scholar 

  37. Liggett, W. H. Jr & Sidransky, D. Role of the p16 tumor suppressor gene in cancer. J. Clin. Oncol. 16, 1197–1206 ( 1998).

    Article  CAS  PubMed  Google Scholar 

  38. Fulci, G. et al. p53 gene mutation and ink4a-arf deletion appear to be two mutually exclusive events in human glioblastoma. Oncogene 19, 3816–3822 ( 2000).

    Article  CAS  PubMed  Google Scholar 

  39. Louis, D. N. The p53 gene and protein in human brain tumors. J. Neuropath. Exp. Neurol. 53, 11–21 ( 1994).

    Article  CAS  PubMed  Google Scholar 

  40. Costello, J. F. et al. Cyclin-dependent kinase 6 (CDK6) amplification in human gliomas identified using two-dimensional separation of genomic DNA. Cancer Res. 57, 1250–1254 ( 1997).

    CAS  PubMed  Google Scholar 

  41. He, J., Reifenberger, G., Liu, L., Collins, V. P. & James, C. D. Analysis of glioma cell lines for amplification and overexpression of MDM2. Genes Chromosomes Cancer 11, 91–96 ( 1994).

    Article  CAS  PubMed  Google Scholar 

  42. von Deimling, A. et al. Subsets of glioblastoma multiforme defined by molecular genetic analysis. Brain Path. 3, 19– 23 (1993).

    Article  CAS  Google Scholar 

  43. van Meyel, D. J. et al. p53 mutation, expression, and DNA ploidy in evolving gliomas: evidence for two pathways of progression. J. Natl Cancer Inst. 86, 1011–1017 ( 1994).

    Article  CAS  PubMed  Google Scholar 

  44. Marutani, M. et al. Dominant-negative mutations of the tumor suppressor p53 relating to early onset of glioblastomamultiforme. Cancer Res. 59, 4765–4769 (1999).

    CAS  PubMed  Google Scholar 

  45. Watanabe, K. et al. Overexpression of the EGF receptor and p53 mutations are mutually exclusive in the evolution of primary and secondary glioblastomas. Brain Pathol. 6, 217–223 (1996).

    Article  CAS  PubMed  Google Scholar 

  46. Li, J. et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 257, 1943–1947 (1997). PubMed

    Article  Google Scholar 

  47. Teng, D. H. et al. MMAC1/PTEN mutations in primary tumor specimens and tumor cell lines. Cancer Res. 57, 5221– 5225 (1997).

    CAS  PubMed  Google Scholar 

  48. Paramio, J. M., Segrelles, C., Casanova, M. L. & Jorcano, J. L. Opposite functions for E2F1 and E2F4 in human epidermal keratinocyte differentiation . J. Biol. Chem. 275, 41219– 41226 (2000).

    Article  CAS  PubMed  Google Scholar 

  49. Hass, R. Retrodifferentiation and cell death. Crit. Rev. Oncol. 5, 359–371 (1994). PubMed

    Article  CAS  Google Scholar 

  50. Hoshimaru, M., Ray, J., Sah, D. W. & Gage, F. H. Dedifferentiation of the immortalized adult neuronal progenitor cell line HC2S2 into neurons by regulatable suppression of the v-myc oncogene. Proc. Natl Acad. Sci. USA 93, 1518–1523 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Prados, M. D. & Levin, V. Biology and treatment of malignant glioma. Semin. Oncol. 27, S1– S10 (2000).

    Google Scholar 

  52. Castro, M. G. et al. Gene therapy strategies for intracranial tumours: glioma and pituitary adenomas. Histol. Histopathol. 15, 1233–1252 (2000).

    CAS  PubMed  Google Scholar 

  53. Pollack, I. F., Okada, H. & Chambers, W. H. Exploitation of immune mechanisms in the treatment of central nervous system cancer. Semin. Pediatr. Neurol. 7, 131–143 (2000).

    Article  CAS  PubMed  Google Scholar 

  54. Finkelstein, S. D. et al. Histological characteristics and expression of acidic and basic fibroblast growth factor genes in intracerebral xenogeneic transplants of human glioma cells. Neurosurgery 34, 136–143 (1994).

    CAS  PubMed  Google Scholar 

  55. Aguzzi, A., Brandner, S., Isenmann, S., Steoinbach, J. P. & Sure, U. Transgenic and gene disrupution techniques in the study of neurocarcinogenesis. Glia 15, 348–364 (1995).

    Article  CAS  PubMed  Google Scholar 

  56. Macleod, K. F. & Jacks, T. Insights into cancer from transgenic mouse models. J. Pathol. 187, 43–60 (1999).A review of the uses of germline-modification strategies in mice for studying human cancers.

    Article  CAS  PubMed  Google Scholar 

  57. Fisher, G. H. et al. Development of a flexible and specific gene delivery system for production of murine tumor models. Oncogene 18, 5253–5260 (1999). A review of the somatic-cell gene-transfer approach to modelling cancer in mice.

    Article  CAS  PubMed  Google Scholar 

  58. Holland, E. C. et al. Astrocytes can give rise to oligodendrocytomas and astrocytomas after transfer of middle T antigen in mice. Am. J. Pathol. 157, 1031–1037 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Chin, L. et al. Essential role for oncogenic Ras in tumour maintenance. Nature 400, 468–472 ( 1999).

    Article  CAS  PubMed  Google Scholar 

  60. Felsher, D. W. & Bishop, J. M. Reversible tumorigenesis by MYC in hematopoietic lineages. Mol. Cell 4, 199–207 (1999).

    Article  CAS  PubMed  Google Scholar 

  61. Pelengaris, S., Littlewood, T., Khan, M., Elia, G. & Evan, G. Reversible activation of c-Myc in skin: induction of a complex neoplastic phenotype by a single oncogenic lesion. Mol. Cell 3 , 565–577 (1999).

    Article  CAS  PubMed  Google Scholar 

  62. Wessenberger, J. et al. Development and malignant progression of astrocytomas in GFAP-v-src transgenic mice. Oncogene 14, 2005–2013 (1997).

    Article  Google Scholar 

  63. Uhrbom, L., Hesselager, G., Nister, M. & Westermark, B. Induction of brain tumors in mice using a recombinant platelet-derived growth factor B-chain retrovirus. Cancer Res. 58, 5275–5279 (1998).

    CAS  PubMed  Google Scholar 

  64. Reilly, K. M., Loisel, D. A., Bronson, R. T., McLaughlin, M. E. & Jacks, T. Nf1; Trp53 mutant mice develop glioblastoma with evidence of strain-specific effects. Nature Genet. 26, 109–113 (2000).This paper reported cooperation between signalling pathways and cell-cycle arrest pathways in the formation of gliomas in a mouse model.

    Article  CAS  PubMed  Google Scholar 

  65. Serrano, M. et al. Role of the INK4a locus in tumor suppression and cell mortality. Cell 85, 27– 37 (1996).

    Article  CAS  PubMed  Google Scholar 

  66. Holland, E. C., Hively, W.P., Gallo, V. & Varmus, H. E. Overexpression of cdk4 but not loss of INK4a-ARF induces hyperploidy in cultured mouse astrocytes, modeling mutations in the G1 arrest pathway in human gliomas . Genes Dev. 12, 3644–3649 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kamijo, T., Bodner, S., van de Kamp, E., Randle, D. H. & Sherr, C. J. Tumor spectrum in ARF-deficient mice. Cancer Res. 59, 2217– 2222 (1999).

    CAS  PubMed  Google Scholar 

  68. Donehower, L. A. et al. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 356, 215–221 (1992).

    Article  CAS  PubMed  Google Scholar 

  69. Yahanda, A. M., Bruner, J. M., Donehower, L. A. & Morrison, R. S. Astrocytes derived from p53-deficient mice provide a multistep in vitro model for development of malignant gliomas. Mol. Cell. Biol. 15, 4249–5259 ( 1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Jacks, T. et al. Effects of an Rb mutation in the mouse. Nature 359, 295–300 (1992).

    Article  CAS  PubMed  Google Scholar 

  71. Wiliams, B. O. et al. Extensive contribution of Rb-deficient cells to adult chimeric mice with limited histopathological consequences. EMBO J. 13, 4251–4259 ( 1994).

    Article  Google Scholar 

  72. Holland, E. C., Hively, W. P., DePinho, R. A. & Aarmus, H. E. A constitutively active epidermal growth factor receptor cooperates with disruption of G1 cell cycle arrest pathways to induce gliomas in mice. Genes Dev. 12, 3675–3685 ( 1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Lal, A. et al. A public database for gene expression in human cancers. Cancer Res. 59, 5403–5407 (1999).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

I would like to thank Greg Fuller and Joseph Celestino for help with the pathology, Chengkai Dai for help with the cell-lineage discussion, and V. K. Rajasekhar for help with the signal-transduction discussion.

Author information

Authors and Affiliations

Authors

Related links

Related links

DATABASE LINKS

GFAP

PDGF

FGF2

EGF

CNTF

EGFR

ERK

JNK

p38

NF1

PI(3)K

PTEN

AKT1

GSK3

mTOR

FHKR

RB

INK4A

p53

CDK4

CDK6

cyclin D1

MDM2

MYC

Myc

Akt

Nf1

Pdgfb

Ink4a

p53

Rb

Egfr

Cdk4

Gfap

PDGFR

VEGFR

FURTHER INFORMATION

Massachusetts General Hospital Brain Tumour Center

Clinical trials information for brain tumours

ENCYCLOPEDIA OF LIFE SCIENCES

Brain cancers

Brain imaging

Glossary

GLIA

The specialized connective tissue of the central nervous system. It is made up of glial cells such as astrocytes, oligodendrocytes and ependymal cells.

ASTROCYTE

One of the three main cell types in the brain, the others being neurons and oligodendrocytes. Astrocytes act as the scaffold that maintains the brain structure and that supports the functions of both neurons and oligodendrocytes.

OLIGODENDROCYTES

One of the three main cell types that make up the brain parenchyma, the other two being neurons and astrocytes. Oligodendrocytes produce myelin, which insulates axons to alter the conduction properties of neurons.

HYPERTROPHY

A pathological increase in the size of cells or the structure that they form.

BRAIN PARENCHYMA

The inner substance of the brain that is composed primarily of neurons, oligodendrocytes, astrocytes and blood vessels.

SUBPIA

The region directly below the pia, the membrane that forms the limiting edge of the brain. Invading glioma cells tend to accumulate in this region to generate one of the classic secondary structures of human gliomas.

ANAPLASTIC

When cells or tissues revert to a more embryonic or undifferentiated form and have an increased capacity to multiply.

GLIAL FIBRILLARY ACIDIC PROTEIN (GFAP).

An intermediate filament protein. The expression of its gene is limited to astrocytes.

PLATELET-DERIVED GROWTH FACTOR (PDGF).

A growth factor that exists as a homodimer or heterodimer of PDGFA and PDGFB chains. This growth factor binds to and activates the PDGF receptor PDGFRA or PDGFRB.

FIBROBLAST GROWTH FACTOR 2 (FGF2).

This ligand binds to the FGF2 receptor, promotes the proliferation of undifferentiated cells and stimulates angiogenesis.

CILLIARY NEUROTROPHIC FACTOR (CNTF).

This ligand binds to the CNTF receptor and promotes oligodendrocyte and astrocyte differentiation.

EPIDERMAL GROWTH FACTOR RECEPTOR (EGFR)

This receptor is bound by epidermal growth factor and the transforming growth factor-α.

GLYCOGEN SYNTHASE KINASE 3 (GSK3).

A kinase involved in several biological processes, including glucose metabolism and signalling through the Wnt pathway. GSK3 also functions downstream of AKT.

MAMMALIAN TARGET OF RAPAMYCIN (mTOR).

A protein that is activated by AKT and which activates ribosomal protein S6 kinase. S6 kinase alters the ability of the ribosome to translate specific mRNAs.

BAD

BAD promotes apoptosis by dimerizing with and inhibiting BCL-2.

BCL-2

BCL-2 inhibits apoptosis by inhibiting caspase activation.

CASPASE

One of a family of proteases that are activated specifically in apoptotic cells.

FORKHEAD TRANSCRIPTION FACTOR (FKHR).

This protein activates cell death. It is inactivated by AKT-dependent phosphorylation, which relocalizes it from the nucleus to the cytoplasm.

PSEUDODIPLOID

DNA content similar to that of a diploid cell.

E2F1

A transcription factor that is bound to RB during G1 cell-cycle arrest and is released after phosphorylation of RB by CDK2 or CDK4. Free E2F1 then alters gene expression to lead to cell-cycle progression.

XENOGRAFT

Cells derived from one species that are implanted into a host of another species; for example, human tumour cells implanted into a mouse.

ALLOGRAFT

Cells implanted into a host that are derived from another individual of the same species.

POLYOMA VIRUS MIDDLE T ANTIGEN

Viral gene product that activates many signalling pathways that are activated by the PDGF receptors.

H-RAS

Harvey Ras. Activated Ras allele initially isolated from Moloney mouse leukaemia virus.

V-SRC

A virally encoded oncogene originally isolated from the Rous sarcoma virus. This gene encodes a deleted and activated version of the cellular Src gene. Expression of this gene activates multiple signalling pathways.

V-ERBB

A virally encoded oncogene originally isolated from the chicken erythroblastosis virus. This gene encodes an activated variant of the EGFR.

V-SIS

A virally encoded oncogene originally isolated from the simian sarcoma virus. This gene encodes the complete sequence of the PDGFB chain. Overexpression results in autocrine stimulation of the PDGF receptor.

K-RAS

Kirsten Ras. Activated Ras allele initially isolated from Kirsten mouse leukaemia virus.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Holland, E. Gliomagenesis: genetic alterations and mouse models. Nat Rev Genet 2, 120–129 (2001). https://doi.org/10.1038/35052535

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/35052535

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing