Elsevier

Seminars in Cancer Biology

Volume 50, June 2018, Pages 32-41
Seminars in Cancer Biology

FOXO family in regulating cancer and metabolism

https://doi.org/10.1016/j.semcancer.2018.01.018Get rights and content

Abstract

FOXO proteins are a sub-group of a superfamily of forkhead box (FOX)-containing transcription factors (TFs). FOXOs play an important role in regulating a plethora of biological activities ranging from development, cell signaling, and tumorigenesis to cell metabolism. Here we mainly focus on reviewing the role of FOXOs in regulating tumor and metabolism. Moreover, how crosstalk among various pathways influences the function of FOXOs will be reviewed. Further, the paradoxical role for FOXOs in controlling the fate of cancer and especially resistance/sensitivity of cancer to the class of drugs that target PI3K/AKT will also be reviewed. Finally, how FOXOs regulate crosstalk between common cancer pathways and cell metabolism pathways, and how these crosstalks affect the fate of the cancer will be discussed.

Introduction

FOXO proteins are a sub-group of a superfamily of forkhead box (FOX)-containing transcription factors (TFs). The founding member of this superfamily, dFOXO, was discovered as the gene whose mutation causes defective development of fruit fly (D. melanogaster), with an extra head (hence forkhead phenotype) [1]. A hallmark of this superfamily of proteins is that they contain a conserved protein domain, forkhead box (FOX), which consists of about 100 amino acid residues that directly bind DNA sequence in the enhancers of various target genes [2]. The FOX domain in the TFs binds DNA with a helix-turn-helix motif with two large loops (like wings), so earlier on these TFs were also referred to as winged helix/forkhead TFs [3]. As a growing number of the TFs with the conserved domain were identified from various species ranging from yeast to humans, surpassing 100, a necessity arose to systematically classify and name these TFs. Phylogenetic analysis of the sequences of the known chordate FOX proteins classifies these TFs into 19 subclasses, ranging from FOXA to FOXS [[3], [4]]. The structure and function of FOXO family of TFs have been excellently reviewed [[5], [6], [7]], and here we mainly focus on reviewing their key role in regulating cancer and the potential crosstalk between cancer and cell metabolism related signaling pathways.

Section snippets

FOXO family members

In invertebrates such as nematode C. elegans and the fruit fly, there is only one FOXO homologous gene, termed daf-16 in the worm and dFOXO in the fly. In mammals, four subfamily members have been identified: FOXO1 (previously also known as FKHR), FOXO3 (aka FKHRL1), FOXO4 (aka AFX) [8] and FOXO6. FOXO1, FOXO3 and FOXO4 mRNAs are expressed ubiquitously in varying levels in mammals [[9], [10]]. Expression of FOXOs has certain tissue preference. FOXO1 is highly expressed in adipose tissues, while

Genetic and biochemistry characters of FOXO proteins

The highly conserved DNA-binding forkhead box domain (∼100-amino-acid Forkhead box) is located in the N-terminal region of the protein, while the transactivation domain is located in the C-terminal region (Fig. 1). In addition, FOXOs except FOXO6 also contain nuclear export sequence and nuclear localization signals that are responsible for shuttling of FOXOs between the nucleus and cytoplasm [12]. FOXO6 lacks NES and its distribution is nuclear independent of external signaling [11]. FOXO

Regulation of FOXOs by PI3K/AKT

A prominent feature of FOXOs is that their cellular localization in the cytoplasm and nucleus is tightly regulated. There is a nuclear localization signal (NLS) domain and a nuclear export signal (NES) domain in FOXOs, facilitating the shuttling of FOXOs between the cytoplasm and the nucleus (Fig. 1). Certain cell signaling events, such as insulin signaling, changes the balance of their cellular distribution. On activation by the extracellular signals such as insulin or insulin-like growth

Other posttranslational modifications (PTMs) of FOXOs

Besides phosphorylation of FOXOs by various kinases to regulate their functions, FOXOs are also subject to other PTMs in various physiological or pathological conditions. These PTMs include ubiquitination, acetylation, and methylation [44], and play a crucial role in regulating FOXOs’ activity and stability.

The role of FOXOs to suppress tumorigenesis

FOXOs have diverse functions and control various biological functions, including cell cycle arrest at the G1-S [57] and G2-M [58] checkpoints, detoxification of reactive oxygen species (ROS) [59], repair of damaged DNA [[58], [60]], and apoptosis [61]. It is well known that high expression or activation of FOXOs in cells is associated with anti-proliferation and apoptosis, functions related to tumor suppression. FOXO3 is well known for its role in repressing cell proliferation and its

Paradoxical role for FOXOs in controlling the fate of cancer

Although FOXOs are generally associated with suppressing cell proliferation and tumorigenesis, in certain conditions, FOXOs are also involved in driving or sustaining tumor cell growth or leading tumor cells to drug resistance. For instance, high expression of FOXO6 is positively correlated with the progression and poor prognosis of gastric cancer [67], and it promotes tumorigenicity via upregulation of C-Myc [65]. In lung cancer cells, inhibition of mutant EGFR triggers SOX2-FOXO6-dependent

FOXOs regulate cell metabolism in multiple organs

As potent effectors of the insulin signaling pathway, FOXOs are involved in the insulin pathway-regulated process of metabolism in different type cells or tissues. In liver, overexpression of FOXO1 inhibits the expression of genes involved in glycolysis, the pentose phosphate pathway and lipogenesis, resulting in increased glucose synthesis under fasting and insulin resistance [80]. Mice with FOXO1- deletion in the liver are resistant to high fat diet-induced insulin resistance [[81], [82]].

The role of FOXOs in regulating beta cells

The maintenance of beta cell function and mass is critical for glucose homeostasis. The previous studies have demonstrated that FOXO1 is involved in regulating beta cell mass and protecting beta cell function [[81], [97], [129], [130]].

The mechanism underlying the role of FOXOs in regulating different cancers

FOXOs are crucial factors for promoting or sustaining a subset of AML [73] and cause drug resistance in HER2+ breast cancer cells [[74], [139]]. Thus, it is interesting to understand whether this mechanism also applies to other types of cancers, especially HER2+ or other PI3 K/AKT pathway-augmented cancers. This is important because many types of cancers, directly or indirectly, harbors much enhanced PI3K/AKT pathways, and numerous efforts are made to develop new drugs to target these pathways

Conflict of interest

None.

Acknowledgements

We acknowledge that this work was supported in part by grants from the NIH (1-R01-CA-178856 and R01 DK097555), AACR-Neuroendocrine tumor research foundation (NETRF), a Harrington Discovery Institute Innovator Scholar award, a CTSA-ITMAT pilot grant at University of Pennsylvania, and an international postdoctoral exchange fellowship program from China Postdoctoral Science Foundation. We apologize for not being able to cite all the important publications due to space limitation.

References (150)

  • J.W. Lee et al.

    Protein kinase A-alpha directly phosphorylates FoxO1 in vascular endothelial cells to regulate expression of vascular cellular adhesion molecule-1 mRNA

    J. Biol. Chem.

    (2011)
  • M.K. Lehtinen et al.

    A conserved MST-FOXO signaling pathway mediates oxidative-stress responses and extends life span

    Cell

    (2006)
  • E.L. Greer et al.

    The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor

    J. Biol. Chem.

    (2007)
  • M.C. Hu et al.

    IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a

    Cell

    (2004)
  • N. Chapuis et al.

    IkappaB kinase overcomes PI3K/Akt and ERK/MAPK to control FOXO3a activity in acute myeloid leukemia

    Blood

    (2010)
  • L.P. van der Heide et al.

    Regulation of FoxO activity by CBP/p300-mediated acetylation

    Trends Biochem. Sci.

    (2005)
  • M.E. Giannakou et al.

    The interaction between FOXO and SIRT1: tipping the balance towards survival

    Trends Cell Biol.

    (2004)
  • M.M. Mihaylova et al.

    Class IIa histone deacetylases are hormone-activated regulators of FOXO and mammalian glucose homeostasis

    Cell

    (2011)
  • Y. Takahashi et al.

    Asymmetric arginine dimethylation determines life span in C. elegans by regulating forkhead transcription factor DAF-16

    Cell Metab.

    (2011)
  • S. Ramaswamy et al.

    A novel mechanism of gene regulation and tumor suppression by the transcription factor FKHR

    Cancer Cell

    (2002)
  • P.F. Dijkers et al.

    Expression of the pro-apoptotic Bcl-2 family member Bim is regulated by the forkhead transcription factor FKHR-L1

    Curr. Biol.

    (2000)
  • J.H. Paik et al.

    FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis

    Cell

    (2007)
  • X. Zhang et al.

    Akt, FoxO and regulation of apoptosis

    Biochim. Biophys. Acta

    (2011)
  • L. Yan et al.

    PP2A regulates the pro-apoptotic activity of FOXO1

    J. Biol. Chem.

    (2008)
  • J. Liu et al.

    FOXO1-FGFR1 fusion and amplification in a solid variant of alveolar rhabdomyosarcoma

    Mod. Pathol.

    (2011)
  • S.M. Sykes et al.

    AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias

    Cell

    (2011)
  • S. Matkar et al.

    An epigenetic pathway regulates sensitivity of breast cancer cells to HER2 inhibition via FOXO/c-Myc Axis

    Cancer Cell

    (2015)
  • C.L. Bigarella et al.

    FOXO3 transcription factor is essential for protecting hematopoietic stem and progenitor cells from oxidative DNA damage

    J. Biol. Chem.

    (2017)
  • W. Zhang et al.

    FoxO1 regulates multiple metabolic pathways in the liver: effects on gluconeogenic, glycolytic, and lipogenic gene expression

    J. Biol. Chem.

    (2006)
  • Y. Kamei et al.

    A forkhead transcription factor FKHR up-regulates lipoprotein lipase expression in skeletal muscle

    FEBS Lett.

    (2003)
  • Y.I. Kitamura et al.

    FoxO1 protects against pancreatic beta cell failure through neuroD and MafA induction

    Cell Metab.

    (2005)
  • C. Postic et al.

    Role of the liver in the control of carbohydrate and lipid homeostasis

    Diabetes Metab.

    (2004)
  • K.H. Kaestner et al.

    Unified nomenclature for the winged helix/forkhead transcription factors

    Genes Dev.

    (2000)
  • B.C. Jackson et al.

    Update of human and mouse forkhead box (FOX) gene families

    Hum. Genom.

    (2010)
  • T. Obsil et al.

    Structure/function relationships underlying regulation of FOXO transcription factors

    Oncogene

    (2008)
  • E.L. Greer et al.

    FOXO transcription factors at the interface between longevity and tumor suppression

    Oncogene

    (2005)
  • J. Corral et al.

    Acute leukemias of different lineages have similar MLL gene fusions encoding related chimeric proteins resulting from chromosomal translocation

    Proc. Natl. Acad. Sci. U. S. A.

    (1993)
  • T. Furuyama et al.

    Identification of the differential distribution patterns of mRNAs and consensus binding sequences for mouse DAF-16 homologues

    Biochem. J

    (2000)
  • G. Rena et al.

    Two novel phosphorylation sites on FKHR that are critical for its nuclear exclusion

    EMBO J.

    (2002)
  • J. Gilley et al.

    FOXO transcription factors directly activate bim gene expression and promote apoptosis in sympathetic neurons

    J. Cell Biol.

    (2003)
  • J. Nakae et al.

    Differential regulation of gene expression by insulin and IGF-1 receptors correlates with phosphorylation of a single amino acid residue in the forkhead transcription factor FKHR

    EMBO J.

    (2000)
  • V. Obsilova et al.

    14-3-3 Protein interacts with nuclear localization sequence of forkhead transcription factor FoxO4

    Biochemistry

    (2005)
  • A. Brunet et al.

    14-3-3 transits to the nucleus and participates in dynamic nucleocytoplasmic transport

    J. Cell Biol.

    (2002)
  • J.M. Kramer et al.

    Expression of Drosophila FOXO regulates growth and can phenocopy starvation

    BMC Dev. Biol.

    (2003)
  • S. Gottlieb et al.

    daf-2, daf-16 and daf-23: genetically interacting genes controlling Dauer formation in Caenorhabditis elegans

    Genetics

    (1994)
  • K.D. Kimura et al.

    daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans

    Science

    (1997)
  • K. Lin et al.

    daf-16: An HNF-3/forkhead family member that can function to double the life-span of Caenorhabditis elegans

    Science

    (1997)
  • A.B. Muhammad et al.

    Menin and PRMT5 suppress GLP1 receptor transcript and PKA-mediated phosphorylation of FOXO1 and CREB

    Am. J. Physiol. Endocrinol. Metab.

    (2017)
  • J. Buteau et al.

    Transcription factor FoxO1 mediates glucagon-like peptide-1 effects on pancreatic beta-cell mass

    Diabetes

    (2006)
  • W.J. Song et al.

    Exendin-4 stimulation of cyclin A2 in beta-cell proliferation

    Diabetes

    (2008)
  • Cited by (75)

    • The pyruvate kinase of the whiteleg shrimp Litopenaeus vannamei: Gene structure and responses to short term hypoxia

      2023, Comparative Biochemistry and Physiology -Part A : Molecular and Integrative Physiology
    View all citing articles on Scopus
    1

    Current address: Division of Oncology and Center for Childhood Cancer Research, The Children’s Hospital of Philadelphia, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA.

    View full text