Elsevier

Ageing Research Reviews

Volume 28, July 2016, Pages 27-36
Ageing Research Reviews

Review
IFI16, an amplifier of DNA-damage response: Role in cellular senescence and aging-associated inflammatory diseases

https://doi.org/10.1016/j.arr.2016.04.002Get rights and content

Highlights

  • Accumulation of senescent cells contributes to aging-related inflammatory diseases in humans.

  • Increased levels of IFI16 protein in human cells contribute to cellular senescence.

  • Expression of IFI16 protein is induced by activation of p53 and IFN-signaling in cells.

  • DNA-damage response induces IFI16 expression through activation of p53 and IFN-signaling.

  • IFI16 acts as an amplifier of DNA-damage response to induce permanent cell cycle arrest.

Abstract

DNA-damage induces a DNA-damage response (DDR) in mammalian cells. The response, depending upon the cell-type and the extent of DNA-damage, ultimately results in cell death or cellular senescence. DDR-induced signaling in cells activates the ATM-p53 and ATM-IKKα/β-interferon (IFN)-β signaling pathways, thus leading to an induction of the p53 and IFN-inducible IFI16 gene. Further, upon DNA-damage, DNA accumulates in the cytoplasm, thereby inducing the IFI16 protein and STING-dependent IFN-β production and activation of the IFI16 inflammasome, resulting in the production of proinflammatory cytokines (e.g., IL-1β and IL-18). Increased expression of IFI16 protein in a variety of cell-types promotes cellular senescence. However, reduced expression of IFI16 in cells promotes cell proliferation. Because expression of the IFI16 gene is induced by activation of DNA-damage response in cells and increased levels of IFI16 protein in cells potentiate the p53-mediated transcriptional activation of genes and p53 and pRb-mediated cell cycle arrest, we discuss how an improved understanding of the role of IFI16 protein in cellular senescence and associated inflammatory secretory phenotype is likely to identify the molecular mechanisms that contribute to the development of aging-associated human inflammatory diseases and a failure to cancer therapy.

Introduction

Exposure of mammalian cells to DNA-damaging agents (e.g., ultraviolet light, ionizing radiations, or cancer chemotherapeutic agents) induces a DNA-damage response (DDR) (Lakin and Jackson, 1999, Bargonetti and Manfredi, 2002, Shiloh, 2006, Shiloh and Ziv, 2013). The response activates certain cell signaling pathways, including well-known ataxia telangiectasia mutated (ATM)-p53 (Kastan, 2007, Shiloh and Ziv, 2013) and recently identified ATM-IKKα/β-interferon (IFN)-β (Yu et al., 2015) pathways. The activation of ATM-p53 pathway in cells stimulates the transcription of the p53 target genes that encode for the inhibitors of cell cycle progression (e.g., p21WAF1/CIP1) (el-Deiry et al., 1993, el-Deiry, 1998). Further, activation of the ATM-IKKα/β-IFN-β signaling pathway in cells increases the expression of the IFN-β (a type I IFN) and its secretion (Yu et al., 2015). Binding of IFN-β to high affinity cell surface receptor in autocrine and paracrine manner activates the JAK/STAT signaling pathway in the target cells (Stark et al., 1998), which stimulates the expression of the IFN-inducible genes that encode for the inhibitors of cell cycle progression and the negative regulators of the telomerase (e.g., IFI16) (Chin et al., 1996, Xin et al., 2003, Xin et al., 2004, Song et al., 2010, Clarke et al., 2010), an enzyme that determines the proliferation potential of cells through maintaining the telomere length (Hahn 2005).

In murine bone marrow-derived macrophages (BMDMs), DNA-damage by ionizing radiations or etoposide treatment releases single and double-stranded DNA into cytoplasm (Härtlova et al., 2015). Upon sensing of cytosolic DNA, the murine structural and functional homologue of the IFI16 protein, p204, recruited the adaptor protein STING to induce the expression of IFN-β through activation of the TBK1 and IRF3. Interestingly, the induction of IFN-β production in BMDMs was dependent upon the ATM kinase. Further, fibroblasts from individuals with Ataxia Telangiectasia (A-T, a premature aging syndrome) also exhibited an increased expression of IFN-β and activation of type I IFN response (an increased expression of the IFN-inducible genes) (Duan et al., 2011a, Duan et al., 2011b). Accordingly, BMDMs from Atm-deficient mice exhibited accumulation of DNA into the cytoplasm (without any treatment with DNA-damaging agents) and activated the type I IFN response (Härtlova et al., 2015).

Depending upon the cell-type and the extent of DNA-damage, activation of DDR-induced signaling in cells ultimately results in either a transient cell cycle arrest, cell death, or permanent cell cycle arrest termed replicative or cellular senescence (Levine, 1997, Bargonetti and Manfredi, 2002, Campisi, 2001, Feng et al., 2008, Shiloh and Ziv, 2013). The p53-mediated induction of p21WAF1/CIP1 protein levels in a variety of cell types (e.g., fibroblasts and epithelial cells) results in a transient cell cycle arrest, thus allowing cells to continue the cell-cycle progression after completion of DNA repair. In contrast, an extensive DNA-damage in cells results in cellular senescence-associated permanent cell cycle arrest (Levine, 1997, Campisi, 2001, Feng et al., 2008, Shiloh and Ziv, 2013).

Cellular senescence (or replicative senescence) is a phenotype, which is accompanied by a failure to re-enter the cell division cycle in response to mitogenic stimulation and by an acquired resistance to oncogenic challenge (Bringold and Serrano, 2000, Campisi, 2001, Campisi, 2013, Salama et al., 2014, Sharpless and Sherr, 2015). In addition to DDR, cellular senescence in human normal cells can be induced by short telomeres (due to an exhaustion of the replication potential), oncogenes (or supra-physiological mitogenic signals), and overexpression of certain tumor suppressor genes (Campisi, 2013, Salama et al., 2014, Sharpless and Sherr, 2015). Further, the activity of both p53 and retinoblastoma tumor suppressor protein (pRb) plays an important role in inducing cellular senescence-associated cell cycle arrest in human normal cells (Bringold and Serrano, 2000, Itahana et al., 2001, Thomas et al., 2003, Feng et al., 2008;). Because senescent cells are unable to proliferate, the induction of cellular senescence in human cells is thought to suppress tumorigenesis (Campisi, 2001, Campisi, 2013, Salama et al., 2014). Although, senescent cells are resistant to oncogenic challenge and do not proliferate, these cells exhibit a secretory phenotype (senescence-associated secretory phenotype or SASP) that is associated with secretion of proinflammatory cytokines and “exososmes” (Lehmann et al., 2008, Hoare and Narita, 2013, Franceschi and Campisi, 2014, Salama et al., 2014). Exosomes (microvesicles, 50–100 nm) contain pathogen-associated and damage-associated molecular patterns (PAMPs and DAMPs), cytokines, a number of proteins (e.g., CD81 and HMGB1), and DNA (up to ∼1000 bp) (Lehmann et al., 2008, Zhu et al., 2014). Notably, SASP is closely associated with aging of muticellular tissues and organs (Zhu et al., 2014). Further, accumulation of senescent cells (and associated SASP) in tissues and organs is thought to contribute to organismal aging and inflammation-associated human diseases, including the development of certain human cancers (Collado et al., 2007, Franceschi and Campisi, 2014, Childs et al., 2015). Notably, accumulation of senescent cells has been reported in tumors after radiation and/or chemotherapy of cancer patients (Salama et al., 2014, Sharpless and Sherr, 2015).

All cell types produce low constitutive levels of IFN-β (Taniguchi and Takaoka, 2001). This low constitutive IFN-β production, which is maintained in part by activation of the IRF7 transcription factor, is important to maintain various physiological functions and regulate signaling of other cytokines (Taniguchi and Takaoka, 2001). However, an increased production of IFN-β in response to viral infections or activation of DDR, plays an important role in antiviral and DNA-damage responses (Stark and Darnell, 2012). Binding of secreted IFN-β to cell surface high affinity receptor (a heterodimer of the IFNAR1 and IFNAR2 subunits) activates the JAK-STAT signaling pathway in IFN-responsive cells (Stark et al., 1998). The pathway activates STAT1 transcription factor through its phosphorylation on Tyr-701 (indicated as PY-STAT1) by JAK kinase. The activated STAT1 and STAT2, along with IRF9, form the phosphorylated ISGF3 (P-ISGF3) transcription factor, which in the nucleus, stimulates the transcription of >2000 IFN-stimulated genes (ISGs) (Der et al., 1998). Proteins encoded by these genes mediate various biological and physiological activities of the type I IFNs. Notably, treatment of cells with the IFN-β also activates DDR through the generation of reactive oxygen species (ROS) that results in an increased expression of p53 (Takaoka et al., 2003) and activation of the ATM-p53 pathway (Moiseeva et al., 2006, Kim et al., 2009, Duan et al., 2011a, Duan et al., 2011b). Activated p53 induces the expression of the p53-responsive genes, including the IFI16 and p21CIP1 (el-Deiry et al., 1993, Song et al., 2008). Increased expression of a set of ISGs, including the IFI16, in a variety of cell types is associated with cellular senescence-associated cell cycle arrest (Fridman and Tainsky, 2008, Purcell et al., 2014). Further, the expression of certain ISGs is up-regulated in fibroblasts from individuals with premature aging syndrome, including Ataxia Telangiectasia (Duan et al., 2011a, Duan et al., 2011b) and Hutchinson-Gilford progeria (progeria) (Sugita et al., 1995).

Although the links between DDR-induced activation of the ATM-p53 and ATM-IKKα/β-IFN-β signaling pathways and the onset of cellular senescence have been known (Shiloh, 2006, Kastan, 2007, Shiloh and Ziv, 2013, Yu et al., 2015), the role of effector proteins whose expression and/or functions are regulated by these signaling pathways is not well understood. In particular, the role of p53 and IFN-inducible IFI16 protein remains unclear in the onset of cellular senescence in vivo, the development of SASP, and aging-associated inflammatory diseases. In this review, we discuss how an improved understanding of the role of IFI16 protein in the amplification of DDR and the onset of cellular senescence could identify the molecular mechanisms that contribute to the development of aging-related inflammatory diseases and a failure to cancer therapy in patients.

Section snippets

IFI16, a member of the IFN-inducible PYHIN protein family

IFI16 protein is a member of the IFN-inducible protein family (Choubey et al., 2008, Choubey et al., 2010). The family includes the murine (e.g., p202, 203, p204, and Aim2 etc.) and human (e.g., IFI16, IFIX, MNDA, and AIM2) proteins. Most proteins (except the p202 protein) in the family share a PYRIN domain (PYD) in the N-terminus and a partially conserved HIN domain comprising ∼200-amino acids in the C-terminus. Therefore, the family is referred as the PYHIN-family (Schattgen and Fitzgerald,

Potentiation of the ATM-p53 signaling by IFI16 protein

ATM protein kinase (encoded by the ATM gene) is a key regulator of the cellular response to DNA double-strand breaks (Lakin and Jackson, 1999, Kastan, 2007, Shiloh and Ziv, 2013). Ataxia telangiectasia in individuals with the ATM gene mutation is characterized by progressive cerebellar ataxia, immunodeficiency, premature aging, and increased cancer incidences (von Zglinicki et al., 2005). Further, cells that are derived from the individuals with AT are defective in activation of p53 upon

Stimulation of IFN-β expression and IFN-signaling by IFI16

Among a number of cytosolic and nuclear DNA sensors identified recently, the IFI16 protein has emerged as an important stimulator of the IFN-β expression in myeloid and non-myeloid cells (Unterholzner et al., 2010, Jakobsen and Paludan, 2014, Thompson et al., 2014, Dutta et al., 2015, Diner et al., 2015). In myeloid cells, upon sensing cytosolic DNA (a “danger” signal), IFI16 protein recruits the adaptor protein STING to activate the TBK1-IRF3 axis to activate the transcription of the IFNB gene

Cellular senescence-associated cell cycle arrest and IFI16

As noted above, replicative or cellular senescence is defined as a failure to re-enter the cell division cycle in response to mitogenic stimulation and by an acquired resistance to oncogenic challenge (Campisi, 2013, Salama et al., 2014, Sharpless and Sherr, 2015). For HDFs in culture, it is defined as a cell cycle arrest that appears beyond 50 ± 10 population doublings and is associated with telomeres’ shortening (Bringold and Serrano, 2000, Campisi, 2001). This senescence-associated cell cycle

IFI16 in aging-associated cancers and inflammation-associated diseases

The loss of IFI16 expression in human breast (Fujiuchi et al., 2004) and prostate (Xin et al., 2003) epithelial cells is associated with the development of cancers. However, IFI16 expression increased in benign prostate hyperplasia (BPH), an aging-associated disease in which senescent cells accumulate in prostate (Xin et al., 2003). Further, increased expression of IFI16 protein in squamous cell carcinoma was associated with keratinocyte differentiation (Imadome et al., 2010) and in ovarian

Conclusions

Senescent cells accumulate as a part of normal aging in humans, during disease-related premature aging, and in response to certain cancer therapy in patients (Salama et al., 2014, Sharpless and Sherr, 2015). Further, the accumulation of senescent cells in humans has emerged as an important contributor to the development of aging-related diseases such as certain cancers. Consequently, cellular senescence has emerged as a potential target for preventing the development of aging-related diseases

Conflict of interest

The authors have declared no conflict of interest.

Acknowledgements

This study was supported in part by a Merit Award (I01BX001133) from the Veterans Administration (VA) to DC. Authors apologize to those colleagues whose research work could not be cited directly due to space limitations.

References (116)

  • W.S. el-Deiry et al.

    WAF1, a potential mediator of p53 tumor suppression

    Cell

    (1993)
  • N. Fujiuchi et al.

    Requirement of IFI16 for the maximal activation of p53 induced by ionizing radiation

    J. Biol. Chem.

    (2004)
  • M.R. Gubbels Bupp

    Sex the aging immune system, and chronic disease

    Cell. Immunol.

    (2015)
  • A. Härtlova et al.

    DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity

    Immunity

    (2015)
  • M.R. Jakobsen et al.

    IFI16: At the interphase between innate DNA sensing and genome regulation

    Cytokine Growth Factor Rev.

    (2014)
  • T. Jin et al.

    Structures of the HIN domain: DNA complexes reveal ligand binding and activation mechanisms of the AIM2 inflammasome and IFI16 receptor

    Immunity

    (2012)
  • M.B. Kastan

    Wild-type p53: tumors can’t stand it

    Cell

    (2007)
  • N. Kerur et al.

    IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi Sarcoma-associated herpesvirus infection

    Cell Host Microbe

    (2011)
  • K.S. Kim et al.

    Interferon-gamma induces cellular senescence through p53-dependent DNA damage signaling in human endothelial cells

    Mech. Ageing Dev.

    (2009)
  • A.J. Levine

    p53: the cellular gatekeeper for growth and division

    Cell

    (1997)
  • J.C. Liao et al.

    Interferon-inducible protein 16: insight into the interaction with tumor suppressor p53

    Structure

    (2011)
  • J. Mazibrada et al.

    In vivo growth inhibition of head and neck squamous cell carcinoma by the interferon-inducible gene IFI16

    Cancer Lett.

    (2010)
  • R. Panchanathan et al.

    Bisphenol A (BPA) stimulates the interferon signaling and activates the inflammasome activity in myeloid cells

    Mol. Cell. Endocrinol.

    (2015)
  • R. Raffaella et al.

    The interferon-inducible IFI16 gene inhibits tube morphogenesis and proliferation of primary, but not HPV16 E6/E7-immortalized human endothelial cells

    Exp. Cell Res.

    (2004)
  • J.F. Schlaak et al.

    Cell-type and donor-specific transcriptional responses to interferon-alpha. Use of customized gene arrays

    J. Biol. Chem.

    (2002)
  • X. Shi et al.

    IFI16 mis-localization can be a contributing factor to hepatocellular carcinoma progression

    Med. Hypotheses

    (2014)
  • Y. Shiloh

    The ATM-mediated DNA-damage response: taking shape

    Trends Biochem. Sci.

    (2006)
  • G.R. Stark et al.

    The JAK-sTAT pathway at twenty

    Immunity

    (2012)
  • K. Sugita et al.

    Reduction of unscheduled DNA synthesis and plasminogen activator activity in Hutchinson-gilford fibroblasts during passaging in vitro: partial correction by interferon-beta

    Mutat. Res.

    (1995)
  • J.A. Aglipay et al.

    A member of the Pyrin family IFI16, is a novel BRCA1-associated protein involved in the p53-mediated apoptosis pathway

    Oncogene

    (2003)
  • F. Alimirah et al.

    IFI16 in human prostate cancer

    Mol. Cancer Res.

    (2007)
  • M.A. Ansari et al.

    Constitutive interferon-inducible protein 16-inflammasome activation during Epstein-barr virus latency I, II, and III in B and epithelial cells

    J. Virol.

    (2013)
  • M.A. Ansari et al.

    Herpesvirus genome recognition induced acetylation of nuclear IFI16 is essential for its cytoplasmic translocation, inflammasome and IFN-β responses

    PLoS Pathog.

    (2015)
  • B. Azzimonti et al.

    Altered patterns of the interferon-inducible gene IFI16 expression in head and neck squamous cell carcinoma: immunohistochemical study including correlation with retinoblastoma protein, human papillomavirus infection and proliferation index

    Histopathology

    (2004)
  • S. Banin et al.

    Enhanced phosphorylation of p53 by ATM in response to DNA damage

    Science

    (1998)
  • J. Bargonetti et al.

    Multiple roles of the tumor suppressor p53

    Curr. Opin. Oncol.

    (2002)
  • M. Bawadekar et al.

    The extracellular IFI16 protein propagates inflammation in endothelial cells via p38 MAPK and NF-κB p65 activation

    J. Interferon Cytokine Res.

    (2015)
  • T. Booiman et al.

    Polymorphism in IFI16 affects CD4(+) T-cell counts in HIV-1 infection

    Int. J. Immunogenet.

    (2014)
  • L.J. Briggs et al.

    Novel properties of the protein kinase CK2-site-regulated nuclear-localization sequence of the interferon-induced nuclear factor IFI 16

    Biochem. J.

    (2001)
  • J. Campisi

    Cellular senescence as a tumor-suppressor mechanism

    Trends Cell Biol.

    (2001)
  • J. Campisi

    Aging cellular senescence, and cancer

    Annu. Rev. Physiol.

    (2013)
  • U. Certa et al.

    High density oligonucleotide array analysis of interferon-alpha2a sensitivity and transcriptional response in melanoma cells

    Br. J. Cancer

    (2001)
  • B.G. Childs et al.

    Cellular senescence in aging and age-related disease: from mechanisms to therapy

    Nat. Med.

    (2015)
  • Y.E. Chin et al.

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

    Science

    (1996)
  • D. Choubey et al.

    Interferon-inducible IFI16 protein in human cancers and autoimmune diseases

    Front. Biosci.

    (2008)
  • D. Choubey et al.

    Interferon-inducible p200-family proteins as novel sensors of cytoplasmic DNA: role in inflammation and autoimmunity

    J. Interferon Cytokine Res.

    (2010)
  • C.J. Clarke et al.

    Critical role of the transcription factor AP-1 for the constitutive and interferon-induced expression of IFI 16

    J. Cell. Biochem.

    (2003)
  • C.J. Clarke et al.

    Inducible activation of IFI 16 results in suppression of telomerase activity, growth suppression and induction of cellular senescence

    J. Cell. Biochem.

    (2010)
  • F. Coppedè

    Premature aging syndrome

    Adv. Exp. Med. Biol.

    (2012)
  • S. Costa et al.

    Redistribution of the nuclear protein IFI16 into the cytoplasm of ultraviolet B-exposed keratinocytes as a mechanism of autoantigen processing

    Br. J. Dermatol.

    (2011)
  • Cited by (50)

    • Cytosolic DNA sensor IFI16 proteins: Potential molecular integrators of interactions among the aging hallmarks

      2022, Ageing Research Reviews
      Citation Excerpt :

      As noted above, these innate immune responses include an increase in production of IFN-β and stimulation of the IFN-β-inducible IFI16 proteins. The IFI16 proteins functionally interact with the p53/p21CIP1 and pRb/E2F axes to activate them (Xin et al., 2003; Xin et al., 2004; Duan et al., 2011b; Choubey and Panchanathan, 2016). Notably, activation of the p53/p21CIP1 and pRb/E2F axes is central to the antagonistic (including cellular senescence) as well as the integrative (including altered intercellular communication) aging hallmarks (López-Otín et al., 2013).

    • The significance of interferon gamma inducible protein 16 (IFI16) expression in drug resistant ovarian cancer cell lines

      2022, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      As the role of IFI16 in regulation of gene expression was already described [27,45,46], it may be possible that isoform A and C can be involved in the regulation of drug resistance genes expression (in contrast to isoform B involved in cellular senescence and immune response). IFI16can be present both in cytoplasmic and nuclear compartments [28,30] and can target both signaling pathways [29,37,42–44] and bind to the promoter sequence of genes [40,41]. Hence, we hypothesize that nuclear localization in PAC-resistant cell line suggests direct gene expression regulation.

    • Biological functions of NLRP3 inflammasome: A therapeutic target in inflammatory bowel disease

      2021, Cytokine and Growth Factor Reviews
      Citation Excerpt :

      Five types of pattern-recognition receptors (PRRs), namely, NLRP1, NLRP3, NLRC4, absent-in-melanoma 2, and pyrin, have been verified to form inflammasomes. However, this tendency of other members of PRRs, such as NLRP6 [20] and IFI16 [21], requires further validation. The pyrin domain-containing 3 (NLRP3) inflammasome, which belongs to the NOD-like receptor family, is the most studied inflammasome closely related to IBD [22].

    • The emerging roles of the MARCH ligases in antiviral innate immunity

      2021, International Journal of Biological Macromolecules
    View all citing articles on Scopus
    View full text