Phosphoinositide-3-kinase inhibition elevates ferritin level resulting depletion of labile iron pool and blocking of glioma cell proliferation

https://doi.org/10.1016/j.bbagen.2018.12.013Get rights and content

Highlights

  • Gliomas contain high endogenous PI3K activity that controls cell proliferation.

  • Inhibition of PI3K regulates ferritin at transcript and translation level.

  • PI3K inhibition results decreased labile iron pool and affects cell proliferation.

  • Silencing of ferritin-H reverses labile iron pool and cell proliferation.

  • This is found only in glioma cells but not in primary astrocytes.

Abstract

Background

Elevated endogenous phosphoinositide-3-kinase (PI3K) activity is critical for cell proliferation in gliomas. Iron availability is one of the essential factors for cell growth and proliferation. However, any relation between PI3K and cellular iron homeostasis has not been understood so far.

Methods

Glioma cells and human primary astrocytes were treated with class I PI3K inhibitors to examine regulation of iron homeostasis components. Regulation of ferritin was detected at mRNA and translational level. Labile iron pool (LIP) and cell proliferation were examined in glioma cells and human primary astrocytes.

Results

Blocking of PI3K activity elevated ferritin level by 6–10 folds in glioma cells by augmenting mRNA expression of ferritin subunits and also by influencing ferritin translation. IRE-IRP interaction was affected due to conversion of IRP1 to cytosolic aconitase that was influenced by increased iron-sulfur scaffold protein iron-sulfur cluster assembly enzyme (ISCU) level. Elevated ferritin sequestered LIP to affect cell proliferation that was reversed in silencing ferritin by siRNAs of ferritin-H and ISCU. Human primary astrocyte with little PI3K activity did not show any change in ferritin level, LIP and cell proliferation by PI3K inhibitors.

Conclusions

PI3K inhibition promotes ferritin synthesis by dual mechanism resulting sequestration of iron to limit its availability for cell proliferation in glioma cells but not in primary astrocytes.

General Significance: This observation establishes a relation between PI3K signalling and iron homeostasis in glioma cells. It also implies that activated PI3K controls ferritin expression to ensure availability of adequate iron required for cell proliferation.

Introduction

The phosphoinositide-3-kinase family of genes encodes lipid and protein kinases to regulate multiple cellular processes including cell survival, cell proliferation, cell cycle progression, angiogenesis, invasion and metastasis [[1], [2], [3]]. PI3Ks are divided into three classes based on their structure and substrate specificity [4]. The class I PI3Ks phosphorylate and activate Akt (also known as PKB) to participate in cell signalling pathways involved in cell proliferation and several other important cellular mechanisms [5,6]. Class IA PI3Ks are diverse in mammals as they have three catalytic p110 isoforms (p110α, p110β, and p110δ; each encoded by a separate gene) and seven regulatory adaptor proteins. The p110γ is the sole class IB PI3K and differs from class IA enzymes in the N-terminus end (lacking p85 binding site) with a p101 or p84 regulatory subunit [7]. Class I PI3Ks are major focus of research as they are coupled to extracellular stimuli and involved in a wide range of cellular processes [[8], [9], [10]]. PI3K phosphorylates the 3’-OH position of the inositol ring of inositol phospholipids to generate phosphatidylinositol-3-phosphate (PI-3-P), phosphatidylinositol-3,4-bisphosphate (PI-3,4-P2), and phosphatidylinositol-3,4,5-trisphosphate (PI-3,4,5-P3) [11]. Phosphorylation and activation of Akt (PKB), a serine/threonine kinase, the key mediator of signalling downstream of PI3 kinase, is principally dependent on the production of PIP3 [12].

Glioblastoma multiforme (GBM) is the most common primary tumour of the central nervous system in adults [13] with limited patient survival and is considered to be among the most lethal cancers [14]. The PI3K pathway is frequently over-activated in GBM due to gain-of-function mutations in the structural gene for p110α (PIK3CA) and by loss of phosphatase and tensin homolog (PTEN), a lipid phosphatase and negative regulator of PI3K signalling [15]. It has been reported that PI3K-Akt signalling is elevated in about 88% of all glioblastomas [15,16]. Similar deregulation of the PI3-kinase signalling pathway through mutation of PIK3CA (p110α) and loss of PTEN has also been detected frequently in glioma cell lines [[17], [18], [19]]. It is well established that inhibition of PI3K signalling affects proliferation in GBM primary cells [20,21].

Role of iron in cell proliferation and cell cycle progression has been well documented in cancer cells including gliomas [22,23]. Cellular iron acquisition and retention of excess iron contribute to tumour initiation and growth [23,24]. Cells acquire iron mainly through transferrin receptor-1 (TfR1) to increase labile iron pool (LIP). However, in astrocytes iron is taken up mainly by divalent metal transporter-1 (DMT1) [25]. From LIP, iron is stored within ferritin [26] and utilized in iron containing proteins and enzymes required for cellular homeostasis. Ferritin molecule contains up to 4000 iron atoms in its mineral core. Cytoplasmic ferritin has two subunits of H– (Ft-H) and L-type (Ft-L) and their ratio varies in different cells. Ft-H is widely considered of having ferroxidase activity that helps in mineralization of iron. Recently, poly-r(C) binding proteins (PCBPs) are identified as iron chaperons for loading iron into ferritin [27]. In response to altered cellular iron pool, both Ft-H and Ft-L are regulated at the translational level [26]. They contain a single iron responsive element (IRE) in their 5’untranslated regions (UTR) [24,26]. In iron depleted cells, iron regulatory proteins (IRP1 and IRP2) are activated to bind IRE and subsequently decrease ferritin translation. IRP1 converts to cytosolic aconitase by gaining iron-sulfur cluster while IRP2 is degraded by proteasomal pathway in higher cellular iron level to promote ferritin translation [28]. The iron-sulfur scaffold protein iron-sulfur cluster assembly enzyme (ISCU) is involved in converting IRP1 to cytosolic aconitase [29]. There are a few reports of ferritin regulation at transcriptional level but its regulation at translational level has been studied extensively [23]. The unique iron exporter ferroportin releases excess iron from cellular iron pool [30]. Intracellular iron pool is maintained for different cellular functions including cell proliferation by coordinated regulation of components involved in iron uptake, iron release and iron storage [23,28].

An iron acquisition phenotype has been reported in number of cancers including glioblastomas [31,32]. Considering the role of iron and PI3K activity in proliferation of cancer cells [22,23], a relation between elevated PI3K and iron homeostasis is expected but has not been addressed so far in any cancer cells including gliomas. Here we report that blocking of PI3K activity by various inhibitors of class I PI3K increases Ft-H and Ft-L synthesis by increasing transcripts of ferritin subunits as well as at translational level by affecting IRE-IRP interaction. Resultant higher ferritin level sequesters iron to decrease LIP resulting inhibition of cell proliferation. These effects are observed only in glioma cells but not in primary astrocytes. This study identifies iron sequestration in ferritin as a novel mechanism by which PI3K inhibition results in affecting cell proliferation in gliomas.

Section snippets

Reagents

p110α subunit specific inhibitor PIK75 (PIK), p110β subunit specific inhibitor TGX221 (TGX), p110γ subunit specific inhibitor AS252424 (AS) and p110δ subunit specific inhibitor IC87114 (IC) were purchased from Cayman Chemicals. LY294002 (Cat no. L9908) and Wortmannin (Cat no. W1628) were from Sigma-Aldrich. Ferritin-L monoclonal antibody (Cat no. ab109373), ferroportin antibody (Cat no. ab85370) and PCBP1 antibody (Cat no. ab74793) were obtained from Abcam. Ft-H (Cat no. 3998),

PI3-kinase inhibition promotes ferritin expression in glioma cells

To determine the influence of endogenously activated PI3 kinase on the expression of iron homeostasis components, C6 rat glioma cells were treated with LY294002 (LY), a well-documented reversible pan inhibitor of PI3K. In response to LY treatment (0–50 μM), a concentration dependent increase in expressions of Ft-L and Ft-H were detected by Western blot analysis (Fig. 1A). We detected up to about 8-fold increased Ft-L and about 6-fold increased Ft-H levels by 50 μM LY treatment. Ft-L protein

Discussion

The current study revealed that inhibition of PI3K activity by specific inhibitors of class I PI3K could elevate ferritin levels in glioma cells resulting sequestration of iron into this cellular iron storage component. Thus iron becomes unavailable for cell proliferation. Silencing of ferritin reversed the LIP and cell proliferation implicating PI3K activity in controlling ferritin expression and iron availability. Interestingly, in primary astrocyte, these PI3K inhibitors showed no influence

Funding

This work was supported by the Department of Biotechnology, India to CKM (BT/PR20394/MED/122/24/2016); by the National Initiative on Glial Cell Research in Health and Disease from the Department of Biotechnology to CKM (BT/PR4005/MED/30/670/2011) and PS (NBRC) (BT/PR5350/MED/30/811/2012). CKM acknowledges ICMR-CAR grant to SCMM; University with potential for excellence (UPE-II) and DST-PURSE program to Jawaharlal Nehru University. PG and PS are supported by fellowships from University Grant

Author statement

PG performed majority of the experiments and contributed to the manuscript writing. PS performed experiment. HSP and PS (NBRC) contributed experiments related to primary astrocytes. CKM conceived, designed, analyzed data and wrote the manuscript. All authors read and approved the final manuscript.

Acknowledgement

We acknowledge Dr. Abhishek Mukherjee for experiments related to Fluorescence Microscope. Authors also appreciate the infrastructural and other supports of Jawaharlal Nehru University.

References (58)

  • D.L. Schonberg et al.

    Preferential iron trafficking characterizes glioblastoma stem-like cells

    Cancer Cell

    (2015)
  • N. Tapryal et al.

    Reactive oxygen species regulate ceruloplasmin by a novel mRNA decay mechanism involving its 3′-untranslated region: Implications in neurodegenerative diseases

    J. Biol. Chem.

    (2009)
  • S. Dev et al.

    Role of extracellular hydrogen peroxide on regulation of iron homeostasis genes in neuronal cell: Implication in iron accumulation

    Free Radic. Biol. Med.

    (2015)
  • N. Tapryal et al.

    Catecholamine stress hormones regulate cellular iron homeostasis by a posttranscriptional mechanism mediated by iron regulatory protein: Implication in energy homeostasis

    J. Biol. Chem.

    (2015)
  • S. Roberts et al.

    Ferritin iron kinetics and protein turnover in K562 cells

    J. Biol. Chem.

    (1988)
  • J. Truty et al.

    Iron Prevents Ferritin turnover in Hepatic Cells

    J. Biol. Chem.

    (2001)
  • S. Epsztejn et al.

    H-ferritin subunit overexpression in erythroid cells reduces the oxidative stress response and induces multidrug resistance properties

    Blood

    (1999)
  • G. Nie et al.

    Overexpression of mitochondrial ferritin causes cytosolic iron depletion and changes cellular iron homeostasis

    Blood

    (2005)
  • J.O. Fuss et al.

    Emerging critical roles of Fe-S clusters in DNA replication and repair

    Biochim. Biophys. Acta

    (2015)
  • C. Legendre et al.

    Iron metabolism: a double-edged sword in the resistance of glioblastoma to therapies

    Trends Endocrinol. Metab.

    (2015)
  • H.M. Lederman et al.

    Deferoxamine: a reversible S-phase inhibitor of human lymphocyte proliferation

    Blood

    (1984)
  • V. Corce et al.

    Recent advances in cancer treatment by iron chelators

    Bioorg. Med. Chem. Lett.

    (2016)
  • B. Vanhaesebroeck et al.

    Synthesis and function of 3-phosphorylated inositol lipids

    Annu. Rev. Biochem.

    (2001)
  • R.J. Shaw et al.

    Ras, PI(3)K and mTOR signalling controls tumour cell growth

    Nature

    (2006)
  • S. Guillard et al.

    Molecular pharmacology of phosphatidylinositol 3-kinase inhibition in human glioma

    Cell Cycle

    (2009)
  • F.M. Foster et al.

    The phosphoinositide (PI) 3-kinase family

    J. Cell Sci.

    (2003)
  • B. Vanhaesebroeck et al.

    The PI3K-PDK1 connection: more than just a road to PKB

    Biochem. J.

    (2000)
  • D.A. Cantrell

    Phosphoinositide 3-kinase signalling pathways

    J. Cell Sci.

    (2001)
  • S. Koyasu

    Role of class IA phosphoinositide 3-kinase in B lymphocyte development and functions

    Biochem. Soc. Trans.

    (2004)
  • Cited by (8)

    • Carbon quantum dots (CQDs) modified ZnO/CdS nanoparticles based fluorescence sensor for highly selective and sensitive detection of Fe(III)

      2020, Spectrochimica Acta - Part A: Molecular and Biomolecular Spectroscopy
      Citation Excerpt :

      However, excessive intake of iron (III) ions can cause aging diseases such as Alzheimer's and other neurogenic diseases, and a lack of these in the body can lead to many diseases, such as anemia and cirrhosis [2–4]. The state of intracellular iron can be reflected by the labile iron pool (LIP) [5]. And the iron has two forms of Fe(II) and Fe(III) in the LIP, when the redox homeostasis of iron is destroyed, it will lead to metabolic disorders, such as oxygen transport and storage, redox, and damage the body's immune mechanism, which cause a variety of diseases [6].

    View all citing articles on Scopus
    View full text