Elsevier

Differentiation

Volume 81, Issue 4, April 2011, Pages 233-242
Differentiation

Small-molecule blocks malignant astrocyte proliferation and induces neuronal gene expression

https://doi.org/10.1016/j.diff.2011.02.005Get rights and content

Abstract

In the central nervous system (CNS), neural stem cells (NSCs) differentiate into neurons, astrocytes, and oligodendrocytes—these cell lineages are considered unidirectional and irreversible under normal conditions. The introduction of a defined set of transcription factors has been shown to directly convert terminally differentiated cells into pluripotent stem cells, reinforcing the notion that preserving cellular identity is an active process. Indeed, recent studies highlight that tumor suppressor genes (TSGs) such as Ink4a/Arf and p53, control the barrier to efficient reprogramming, leaving open the question whether the same TSGs function to maintain the differentiated state. During malignancy or following brain injury, mature astrocytes have been reported to re-express neuronal genes and re-gain neurogenic potential to a certain degree, yet few studies have addressed the underlying mechanisms due to a limited number of cellular models or tools to probe this process. Here, we use a synthetic small-molecule (isoxazole) to demonstrate that highly malignant EGFRvIII-expressing Ink4a/Arf−/−; Pten−/− astrocytes downregulated their astrocyte character, re-entered the cell cycle, and upregulated neuronal gene expression. As a collateral discovery, isoxazole small-molecules blocked tumor cell proliferation in vitro, a phenotype likely coupled to activation of neuronal gene expression. Similarly, histone deacetylase inhibitors induced neuronal gene expression and morphologic changes associated with the neuronal phenotype, suggesting the involvement of epigenetic-mediated gene activation. Our study assesses the contribution of specific genetic pathways underlying the de-differentiation potential of astrocytes and uncovers a novel pharmacological tool to explore astrocyte plasticity, which may bring insight to reprogramming and anti-tumor strategies.

Introduction

It is well established that mature astrocytes lack neurogenic potential, especially during late postnatal stages and in adult brain (Costa et al., 2010). Elegant work using genetic fate-mapping strategies confirmed that mature cortical astrocytes are largely quiescent and non-neurogenic, but retain the ability to proliferate and upregulate GFAP and other classical markers of reactive glia (Buffo et al., 2008, Buffo et al., 2005). Despite a few studies that examine the ability of glia to give rise to neuronal cells, this appears to only occur by forced expression of neuronal transcription factors in vitro, while the potential for this is drastically reduced in vivo, likely due to the non-neurogenic microenvironment (Blum et al., 2010, Heins et al., 2002, Berninger et al., 2007). Moreover, the finding that brain tumors often contain a mixture of neuronal and glial cell types has raised the notion that these tumors either contain multipotent or restricted stem/progenitors, or, arise from de-differentiated mature cell types, such as astrocytes (Stiles and Rowitch, 2008). Clearly, there is an urgent need to understand the cellular and molecular mechanisms underlying the proliferation and de-differentiation potential of mature astrocytes. Understanding this process, and especially, developing new strategies or tools to explore the extent of astrocyte plasticity may be relevant for designing neuroregenerative approaches and treating brain tumors.

Recent work from multiple labs indicates that reprogramming to pluripotent stem cells is markedly enhanced with the loss of tumor suppressor genes Ink4a or p53 (Li et al., 2009, Utikal et al., 2009). These data reinforce the connection between maintaining the differentiated state and initiating tumorigenesis. The Ink4a/Arf locus encodes two key tumor suppressor proteins (p16Ink4a and p19Arf) that, respectively, engage two critical anti-proliferative pathways, the retinoblastoma (Rb) and p53 pathways, both important for G1 checkpoint control (Sharpless, 2005, Serrano et al., 1993). Ink4a (as well as its other Ink4 orthologs, Ink4b, Ink4c, and Ink4d) bind and inhibit the D-type cyclin-dependent kinases Cdk4 and Cdk6 that, in turn, relieve the cell-cycle inhibitory activity of Rb. On the other hand, Arf binds to and inactivates the Mdm2 protein, which is an E3 ubiquitin ligase that destabilizes p53. Both expression of p16Ink4a and p19ARF are critical for effective tumor suppression—including GBM, which frequently harbors homozygous deletions of the Ink4a/Arf locus (Hall and Peters, 1996, Kamb et al., 1994, Ueki et al., 1996). Indeed, our previous studies indicate that Ink4a/Arf/ astrocytes can undergo de-differentiation to a stem-like glioma cell and re-express progenitor markers such as Nestin and A2B5, retaining a capacity to become differentiated glial and neuronal progeny (Bachoo et al., 2002).

Several key questions are raised by these studies: (1) are there specific tumor suppressor genes and/or oncogenes that govern the differentiation potential of malignant astrocytes, and (2) what is the extent of phenotypic plasticity of malignant astrocytes and is it reversible? In this report, we use a synthetic small-molecule 3,5-disubstituted isoxazole (compound 1), identified in a previous high-throughput chemical compound screen for inducers of differentiation of P19 embryonal carcinoma cells (Sadek et al., 2008, Schneider et al., 2008), to interrogate the molecular pathways that control the lineage plasticity of malignant astrocytes. We demonstrate that Ink4a/Arf, Pten, and EGFRvIII pathways interact to maintain the differentiated state of astrocytes, and that in this context isoxazole acts as a stem cell modulator (SCM) to trigger neuronal gene expression and block tumor cell proliferation. Our findings provide novel insights into Ink4a/Arf-mediated de-differentiation of malignant astrocytes and identify a potential starting point for future glioma therapeutic drug design. Most importantly, we demonstrate the use of a novel pharmacological tool to explore the phenotypic plasticity of astrocytes, which is relevant in the context of cellular de-differentiation, reprogramming, and malignancy.

Section snippets

Astrocyte cell culture

SS05 cells or primary astrocytes were isolated from cerebral cortices of 5-day-old wild-type, Ink4a/Arff/f, Ptenf/f; Ink4a/Arf/; Ptenf/f, or p53/; Ptenf/f pups according to previous methods (Bachoo et al., 2002). The floxed Ink4a/Arf or Pten allele was deleted using an adenovirus expressing Cre. Infection of astrocytes with lentiviruses expressing constitutively active EGFR (EGFRvIII) has previously been described (Bachoo et al., 2002). Cells were cultured in 10% FBS in DMEM:F12 media

Isoxazole SCMs block cell proliferation and induce neuronal gene expression in malignant astrocytes

To explore the lineage potential of malignant astrocyte cultures, we used a continuous line of cultured astrocytes (SS05) derived from genetically engineered mice with homozygous deletion of both Ink4a/Arf and Pten tumor suppressor genes. SS05 cells initially expressed the astrocyte marker (GFAP), but downregulated their astrocyte phenotype and increased proliferation during in vitro cell culture with 10% FBS (data not shown). SS05 cells also harbor constitutively active epidermal growth factor

Discussion

One of the most surprising and exciting discoveries in neural developmental biology this past decade is the finding that NSCs possess characteristics of glial identity (Goldman, 2003, Mori et al., 2005). Radial glia give rise to neurons in the developing telecenphalon across vertebrate species and radial-like stem cells in the postnatal hippocampus and lateral ventricle subependymal zone differentiate into neurons throughout life (Kriegstein and Alvarez-Buylla, 2009). Why do some glial cells

Acknowledgements

We thank Yan Jiang, Yan Wu, and HK Jeong for technical assistance and Jose Cabrera for artwork. We also thank Gerard Evan, Sarah Comerford, Chun-li Zhang, and Ondine Cleaver for helpful comments on the manuscript. This work was funded by grants from the Cancer Prevention & Research Institute of Texas (RP100674, J.H.), the Welch Foundation (I-1660, J.H.), and National Institutes of Health/National Institute on Aging (AG032383, J.H.).

References (46)

  • C.D. Stiles et al.

    Glioma stem cells: a midterm exam

    Neuron

    (2008)
  • N.E. Sharpless

    INK4a/ARF: a multifunctional tumor suppressor locus

    Mutat. Res.

    (2005)
  • J. Silva et al.

    Nanog is the gateway to the pluripotent ground state

    Cell

    (2009)
  • K. Takahashi et al.

    Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors

    Cell

    (2006)
  • A. Buffo et al.

    Origin and progeny of reactive gliosis: a source of multipotent cells in the injured brain

    Proc. Natl. Acad. Sci. USA

    (2008)
  • A. Buffo et al.

    Expression pattern of the transcription factor Olig2 in response to brain injuries: implications for neuronal repair

    Proc. Natl. Acad. Sci. USA

    (2005)
  • Blum, R., Heinrich, C., Sanchez, R., et al. Neuronal network formation from reprogrammed early postnatal rat cortical...
  • B. Berninger et al.

    Functional properties of neurons derived from in vitro reprogrammed postnatal astroglia

    J. Neurosci.

    (2007)
  • A. Banito et al.

    Senescence impairs successful reprogramming to pluripotent stem cells

    Genes Dev.

    (2009)
  • S.W. Bruggeman et al.

    Ink4a and Arf differentially affect cell proliferation and neural stem cell self-renewal in Bmi1-deficient mice

    Genes Dev.

    (2005)
  • F.B. Furnari et al.

    Malignant astrocytic glioma: genetics, biology, and paths to treatment

    Genes Dev.

    (2007)
  • M. Grunstein

    Histone acetylation in chromatin structure and transcription

    Nature

    (1997)
  • N. Heins et al.

    Glial cells generate neurons: the role of the transcription factor Pax6

    Nat. Neurosci.

    (2002)
  • Cited by (28)

    • Rapid and Efficient Conversion of Human Fibroblasts into Functional Neurons by Small Molecules

      2019, Stem Cell Reports
      Citation Excerpt :

      MEF2 family members are identified as important regulators in modulating neurite growth of TUJ1-expressing neurons in vitro and embryonic neural stem cell differentiation in vivo in the developing nervous system (Lam and Chawla, 2007). In combination with the positive impacts of ISX9 on the other cell systems (Dioum et al., 2011; Zhang et al., 2011), additional studies are necessary to clarify the specific mechanisms of small-molecule-induced reprogramming from fibroblasts to neurons. Although many reports have demonstrated that small molecules can convert one type of terminally differentiated somatic cell to another fully differentiated cell type, there are still various major hurdles ahead that must be overcome.

    • Small Molecules Efficiently Reprogram Human Astroglial Cells into Functional Neurons

      2015, Cell Stem Cell
      Citation Excerpt :

      The successful reprogramming of human astrocytes into functional neurons with chemically synthesized compounds may potentially lead to a novel drug therapy for brain repair. To identify the small molecules for astrocyte-neuron reprogramming, we searched for small molecules that play crucial roles in neurodevelopment and neurodifferentiation (Chambers et al., 2009, 2012; Huangfu et al., 2008; Ladewig et al., 2012; Li et al., 2011; Liu et al., 2013; Sirko et al., 2013; Zhang et al., 2011). We tested a variety of small molecules (20 in total) targeting signaling pathways critical for neurodevelopment, including Noggin, BMP, TGFβ, GSK3β, Wnt, RA, Notch, SHH, cAMP, DNA methylation, and histone deacetylation or methylation.

    • Regulation of inflammation by extracellular acidification and proton-sensing GPCRs

      2013, Cellular Signalling
      Citation Excerpt :

      Isx drive cardiomyogenic and pro-survival transcriptional programs in myocardial infarction model [124,125] and promote neurogenesis in young mice in vivo [121]. The small molecules also inhibit proliferation and induce neuronal gene expression in malignant astrocytes [126] and stimulate insulin production [127]. Although the possibility cannot be ruled out that Isx act also on signaling molecules or receptors other than OGR1, this drug is useful for further characterization of the role of OGR1 in biological actions.

    • Nucleoside diphosphate kinase Nm23-M1 involves in oligodendroglial versus neuronal cell fate decision in vitro

      2012, Differentiation
      Citation Excerpt :

      The gliogenesis therefore could be initiated when the proneural machinery was inhibited (Pforte et al., 2005). In adult, the glial progenitor cells were recently found to be able to produce neurons (Berninger, 2009; Lu et al., 2010; Noctor et al., 2001; Zhang et al., 2011). Additionally, in vitro studies indicated that the glial cells could be differentiated and further cause cell fate change (Kondo and Raff, 2000, 2004).

    View all citing articles on Scopus
    1

    Equal-author contribution.

    View full text