Inhibition of neurosphere proliferation by IFNγ but not IFNβ is coupled to neuronal differentiation

https://doi.org/10.1016/j.jneuroim.2008.10.009Get rights and content

Abstract

Interferons are produced following neural damage as part of the inflammatory response and may thus affect neural stem cell function. We compared the effects of interferon-γ and interferon-β on the proliferation and differentiation of adult murine neural progenitors. Both interferons inhibited neurosphere proliferation due to cell cycle arrest in G1 but only interferon-γ induced neuronal differentiation. Both interferons induced differential phosphorylation of STAT proteins and a modest and late upregulation of the cell cycle regulator p27 but not several other likely cell cycle regulators. Thus in neural progenitor cells, anti-proliferative effects of interferons are not necessarily linked to differentiation.

Introduction

Interferon-β (IFNβ) and interferon γ (IFNγ) belong to a group of cytokines that were first discovered as antiviral agents, but are now known to have other properties including immunomodulation, inhibition of proliferation, regulation of differentiation, and anti-tumor activity (Kalvakolanu, 2000, Pestka et al., 1987, Sen, 2001). In brain injury associated with most neurological disorders, interferons are produced at or near injured sites during inflammation. Stem cell therapy has been proposed as a tool to repair and regenerate the CNS but the success of such therapy is hampered by the limited ability of stem cells to survive, proliferate, migrate or differentiate into neurons in the injured adult CNS.

IFNγ (Type II interferon) is one of the inflammatory cytokines produced following brain injury and it is known to exacerbate the symptoms of Multiple Sclerosis. However, it has been shown to exhibit neurotrophic effects (Chang et al., 1990, Turnley et al., 2001) and promote embryonic neural differentiation, neuronal survival, and neurite outgrowth (Barish et al., 1991). In contrast to IFNγ, IFNβ (Type I interferon) is known as an anti-inflammatory cytokine and immunoregulator and is produced by immune and non-immune cells following microbial infection or inflammatory activation. IFNβ has pleiotropic activities in the CNS, some of which may account for its therapeutic effects in Multiple Sclerosis. These include the inhibition of expression of class II major histocompatibility complex induced by IFNγ, modulation of matrix metalloproteinase activities and interleukin-1 and interleukin-1 receptor antagonist expression (Liu et al., 1998, Ransohoff et al., 1991, Yong et al., 1998). It is also reported to reduce blood-brain barrier permeability (Stone et al., 1995) and T cell infiltration to the CNS (Stuve et al., 1996) and to antagonise IFNγ to downregulate expression of cytokines and iNOS in astrocytes (Hua et al., 1998). IFNβ has potent anti-proliferative effects on tumor cells and modulates the activity of immune cells such as microglia, T cells, macrophages and dendritic cells, but its effects on neural stem cells remain to be elucidated.

Both types of IFNs are known to exert their biological effects via the Janus kinase (JAK)/Signal transducer and activator of transcription (STAT) signaling pathway and the mechanisms of their signal transduction, including in the nervous system, have been extensively reviewed elsewhere (Campbell, 2005, Platanias, 2005). Various STAT members and signaling pathways have been shown to play an important role in the anti-proliferative effect of interferons. Cells treated with IFNβ or IFNγ typically exhibit inhibition of cell cycle progression, either in the form of S phase accumulation in carcinoma cells (Garrison et al., 1996, Kaynor et al., 2002, Murata et al., 2006, Qin et al., 1997, van Koetsveld et al., 2006, Vannucchi et al., 2000), G0/G1 arrest (Verma et al., 2002) or G2 arrest (Vivo et al., 2001). Such effects have been demonstrated mainly in carcinoma cells and the molecular mechanism is due to one or more of the following: a) induction of cyclin-dependent kinase inhibitor (cdk) p21 or p27 (Giandomenico et al., 1998, Kuniyasu et al., 1997), b) activation of p38 MAPK signalling (Verma et al., 2002), c) downregulation of cyclin E and cyclin A expression and activity mediated by STAT1 (Kortylewski et al., 2004). A link between STAT signalling and cell cycle control has been established, with reports that the production of p21 is increased by IFNγ-induced STAT1 (Chen et al., 2000, Chin et al., 1996, Kominsky et al., 1998) to induce growth arrest. STAT3 also appears to be another important player in cell cycle control, since it upregulates cyclins D and A, and downregulates p21 and p27 (Fukada et al., 1998) to promote cell cycle progression. However, in myeloid derived cells stimulated with IL-6, the activated STAT3 induced G1 arrest (Minami et al., 1996, Nakajima et al., 1996). In glioblastoma cells, activation of STAT3 blocked cell cycle progression and abnormal cell proliferation by activating p21 expression (Barre et al., 2003). Therefore, the effects of IFNs on cell cycle progression appear to be cell-type specific.

Given that IFNγ has inflammatory properties and we found previously that it inhibits the proliferation of neurospheres, promotes neuronal differentiation and neurite outgrowth (Wong et al., 2004), here we investigated whether anti-inflammatory IFNβ plays a similar role in these cells and compared the downstream signaling events activated by IFNβ and IFNγ to block proliferation. IFNβ inhibited neurosphere proliferation but had no effect on neuronal differentiation and neurite outgrowth. The anti-proliferative effect of both IFNs was associated with activation of STAT proteins and cell cycle arrest with modest upregulation of p27.

Section snippets

Cell culture and reagents

Neurospheres were isolated from the subventricular zone of adult C57BL/6 mice and cultured as previously described (Turnley et al., 2002) in neurosphere proliferation media consisting of Dulbecco's modified Eagle's medium:F12 (Gibco) containing 0.6% glucose, 13.4 mM NaHCO3, 5 mM HEPES, 100 U/ml penicillin/streptomycin (Gibco BRL) supplemented with 20 ng/ml epidermal growth factor (EGF; BD Bioscience), 10 ng/ml fibroblast growth factor 2 (FGF2; Roche), 0.1 mg/ml apo-transferrin, 25 µg/ml

IFNβ inhibits proliferation of neurospheres similarly to IFNγ and both IFNs induce G1 cell cycle accumulation

To determine whether IFNβ had an effect on neurosphere proliferation as we showed previously for IFNγ (Wong et al., 2004), we treated cells with various doses of IFNβ for up to 7 days and scored the number of viable cells daily from day 4 onwards. A significant reduction in live cell numbers induced by all doses of IFNβ (10 U/ml-5000 U/ml) was observed from day 6 onwards compared to the untreated control cells (1 way ANOVA, p < 0.001, F(5,12) = 126.3)(Fig. 1A). The decrease in cell numbers due to

Discussion

We have shown here that both IFNβ and IFNγ are anti-proliferative for neural progenitor cells. Whilst the decreased proliferation by IFNγ was linked to increased neuronal differentiation and neurite outgrowth, this was not the case for IFNβ.

IFNβ promoted cell cycle arrest of proliferating progenitors, as well as some longer term toxicity. Its anti-proliferative effect is consistent with that reported in carcinoma cells (Johns et al., 1992, Murata et al., 2006, Vannucchi et al., 2000, Vitale et

Acknowledgements

This work was supported by Bayer Healthcare Pharmaceuticals, (project # NSV-015) and the National Health and Medical Research Council of Australia (project 454384). AMT is a NH&MRC Senior Research Fellow.

References (44)

  • ChinY.E. et al.

    Cell growth arrest and induction of cyclin-dependent kinase inhibitor p21 WAF1/CIP1 mediated by STAT1

    Science

    (1996)
  • de KoningJ.P. et al.

    STAT3-mediated differentiation and survival and of myeloid cells in response to granulocyte colony-stimulating factor: role for the cyclin-dependent kinase inhibitor p27(Kip1)

    Oncogene

    (2000)
  • DuttonR. et al.

    Regulation of spinal motoneuron differentiation by the combined action of Sonic hedgehog and neurotrophin 3

    Clin. Exp. Pharmacol. Physiol.

    (1999)
  • DuttonR. et al.

    Sonic hedgehog promotes neuronal differentiation of murine spinal cord precursors and collaborates with neurotrophin 3 to induce Islet-1

    J. Neurosci.

    (1999)
  • FarahM.H. et al.

    Generation of neurons by transient expression of neural bHLH proteins in mammalian cells

    Development

    (2000)
  • FukadaT. et al.

    STAT3 orchestrates contradictory signals in cytokine-induced G1 to S cell-cycle transition

    Embo J.

    (1998)
  • GalderisiU. et al.

    Cell cycle regulation and neural differentiation

    Oncogene

    (2003)
  • GarrisonJ.I. et al.

    Interferon-beta inhibits proliferation and progression through S phase of the cell cycle in five glioma cell lines

    J. Neuro-oncol.

    (1996)
  • GiandomenicoV. et al.

    Apoptosis and growth inhibition of squamous carcinoma cells treated with interferon-alpha, IFN-beta and retinoic acid are associated with induction of the cyclin-dependent kinase inhibitor p21

    Eur. Cytokine Netw.

    (1998)
  • HarvatB.L. et al.

    The role of p27Kip1 in gamma interferon-mediated growth arrest of mammary epithelial cells and related defects in mammary carcinoma cells

    Oncogene

    (1997)
  • HauserP.J. et al.

    STAT3 activation accompanies keratinocyte differentiation

    Cell Growth Differ.

    (1998)
  • HuaL.L. et al.

    Selective inhibition of human glial inducible nitric oxide synthase by interferon-beta: implications for multiple sclerosis

    Ann. Neurol.

    (1998)
  • Cited by (26)

    • Daily exercise improves memory, stimulates hippocampal neurogenesis and modulates immune and neuroimmune cytokines in aging rats

      2013, Brain, Behavior, and Immunity
      Citation Excerpt :

      For example, systemic or central bacterial lipopolysaccharide (LPS) injections activate microglia, potently block neuronal differentiation (Ekdahl et al., 2003; Monje et al., 2003) and disrupt the integration of young neurons into existing hippocampal circuitry (Belarbi et al., 2012). Of the cytokines known to be stimulated by LPS (see Erickson and Banks, 2011), only a handful have been shown to affect in vivo or in vitro neurogenesis (Ben-Hur et al., 2003; Buckwalter et al., 2006; Grotendorst et al., 1989; Liu et al., 2009; Lum et al., 2009; Monje et al., 2003; Qin et al., 2008; Turrin et al., 2001; Vallieres et al., 2002; Villeda et al., 2011). In humans, experimental LPS impairs verbal and non-verbal memory (Reichenberg et al., 2001), but confirming its effects on neurogenesis awaits technology that permits the visualization of neurogenesis in the living brain.

    • Interferon β-1b directly modulates human neural stem/progenitor cell fate

      2011, Brain Research
      Citation Excerpt :

      Therefore, identifying how IFN-β influences NPC physiology is of crucial importance in elucidating the potential benefits of directing therapies that increase IFN-β levels in the CNS. Preliminary studies have suggested that IFN-β promotes NPC differentiation and proliferation but are limited to murine cell models (Hirsch et al., 2009; Lum et al., 2009; Wellen et al., 2009). In this study, we investigate the direct effects of IFN β-1b (Betaseron®) on human NPCs in vitro.

    • Regulation of adult neural precursor cell migration

      2011, Neurochemistry International
      Citation Excerpt :

      This partially involved CCL2/CCR2 as migration in the absence of CCL2 or CCR2 was decreased. IFNγ is also a potent inhibitor of NPC proliferation that promotes neuronal differentiation (Wong et al., 2004; Lum et al., 2009). An intriguing possibility is that IFNγ, in concert with chemokines, may be responsible for the rapid and significant migration of NPCs/neuroblasts to the site of ischemic stroke damage (Arvidsson et al., 2002).

    • Minocycline promotes remyelination in aggregating rat brain cell cultures after interferon-γ plus lipopolysaccharide-induced demyelination

      2011, Neuroscience
      Citation Excerpt :

      In the present study, the increased number of NG2- and O4-positive precursors were not due to cell proliferation, unlike observations in a model of antibody-mediated demyelination (Harrer et al., 2009). The decreased rate of thymidine incorporation observed after the treatment with the inflammatory agents is consistent with other studies showing that IFN-γ and LPS decreased the proliferation of neural stem cells and OPCs (Agresti et al., 1996; Ben-Hur et al., 2003; Lum et al., 2009). In the present culture system at advanced developmental stage, the population of nestin positive stem cells is larger than the populations of NG2 positive and O4 positive cells (data not shown).

    View all citing articles on Scopus
    View full text