Review
GDF15, an emerging key player in human aging

https://doi.org/10.1016/j.arr.2022.101569Get rights and content

Highlights

  • GDF15 is one of the most up-regulated proteins during aging and a key molecule in the mechanisms of human stress response.

  • The role of GDF15 in aging and age-associated diseases is more complex than previously thought and not yet totally clear.

  • GDF15 is proposed to be the central core of a bow tie biological model.

  • GDF15 may play a role in a dormancy program to mediate tissue tolerance during inflammation and tissue injury.

  • GDF15 is associated with both positive and detrimental effects and may be an example of antagonistic pleiotropy.

Abstract

Growth differentiation factor 15 (GDF15) is recently emerging not only as a stress-related mitokine, but also as a key player in the aging process, being one of the most up-regulated protein with age and associated with a variety of age-related diseases (ARDs). Many data indicate that GDF15 has protective roles in several tissues during different stress and aging, thus playing a beneficial role in apparent contrast with the observed association with many ARDs. A possible detrimental role for this protein is then hypothesized to emerge with age. Therefore, GDF15 can be considered as a pleiotropic factor with beneficial activities that can turn detrimental in old age possibly when it is chronically elevated. In this review, we summarize the current knowledge on the biology of GDF15 during aging. We also propose GDF15 as a part of a dormancy program, where it may play a role as a mediator of defense processes aimed to protect from inflammatory damage and other stresses, according to the life history theory.

Introduction

Growth differentiation factor 15 (GDF15), originally described as macrophage inhibitory cytokine-1, is a distant member of the transforming growth factor-β superfamily (Bootcov et al., 1997) and is considered a stress responsive cytokine induced by mitochondrial dysfunction, cellular stress, inflammation or mitochondrial unfolded protein response (UPRmt) pathway, with positive effects on health and lifespan of model organisms. For this reason, it perfectly suits the definition of “mitokine” as provided by Dillin and co-workers (Durieux et al., 2011).

In contrast with this tenet, GDF15 expression is very low in healthy individuals and young subjects (Fujita et al., 2016, Tsai et al., 2018, Conte et al., 2019, Conte et al., 2020a). The levels of GDF15 dramatically increase in chronic or acute illness conditions, in presence of age-related diseases, such as cardiovascular diseases, insulin resistance and type 2 diabetes, neurodegeneration, renal chronic disease and cancer, as well as with aging independently from the health state, thus GDF15 has been proposed as a novel biomarker of biological aging in humans (Lee et al., 2021, Liu et al., 2021). Accordingly, several studies in humans have in fact reported that GDF15 is one of the most up-regulated protein during aging (Fujita et al., 2016; Tsai et al., 2018; Tanaka et al., 2018; Lehallier et al., 2019; Conte et al., 2019, Conte et al., 2020a; Liu et al., 2021). The value of GDF15 as a marker of biological age has been further supported by a recent study reporting that GDF15 is expressed more in subjects with accelerated aging such as Down Syndrome persons, with respect to their siblings of similar age (Conte et al., 2019).

GDF15 is expressed in different types of tissues, such as placenta, skeletal muscle, brain, liver, heart, and it is found at circulating level, however, its pathophysiological role in the aging process is still poorly understood, as several studies suggested opposite functions for this protein. In particular, it is not yet clear whether increased levels of this mitokine in old age have detrimental or beneficial/protective effects on the organisms.

Many studies have proposed an anti-inflammatory role for GDF15. As an example, it has been demonstrated that GDF15 reduces the expression of pro-inflammatory cytokines and prevents the activation of T cells in liver of mice with fibrosis. In addition, GDF15 knockout (KO) induces in mice the production of TNF-α by T cells and the activation of CD4+ and CD8+ T cells, thus aggravating liver injury and fibrosis (Chung et al., 2017). In agreement, Abulizi and co-workers reported that GDF15 KO mice display elevated intensities of inflammatory responses and severe renal and cardiac damage in response to lipopolysaccharide (LPS), while GDF15 transgenic mice are protected from LPS-mediated organ injury (Abulizi et al., 2017). Moreover, an in vitro study on macrophages showed that GDF15 limits the action of TNF-α. These studies are in line with another finding in human prostate, where an inverse correlation between GDF15 levels, quantified by immunohistochemistry image analysis, and CD3+ , CD4+ , CD8+ , CD68+ and INOS+ leukocytes has been demonstrated. Moreover, the expression of IL-8 is downregulated by GDF15 (Lambert et al., 2015). In agreement with these data, a recent study by Moon and colleagues identified a link between age-related induction of GDF15 and the protection of tissues from inflammation, in both humans and mouse models. In particular, these authors demonstrated that aged GDF15 KO mice presented higher levels of inflammatory markers in liver and adipose tissues, as well as impaired glucose homeostasis. Therefore, these data suggest that GDF15 is indispensable for attenuating aging-mediated local and systemic inflammation (Moon et al., 2020).

Similarly to inflammation, there are evidences indicating that GDF15 can have also anti-cancer properties, suggesting a tumor suppressor role for this protein. In a recent in vitro study, the overexpression of GDF15 in lung adenocarcinoma A549 cells inhibits the proliferation, migration and invasion of these cells through the activation of TGF-β/Smad signaling pathway (Duan et al., 2019). In agreement, another study indicates that the overexpression of GDF15 significantly suppresses non-small-cell lung cancer cell proliferation and induces apoptosis (Lu et a, 2017). Moreover, GDF15 overexpressing mice are protected from prostate cancer growth, thanks to the activation of CD8+ cytotoxic lymphocytes (Husaini et al., 2020).

In contrast to the above described protective role of GDF15, many other studies indicate that GDF15 can also play a pro-inflammatory role. For example, a study indicates that GDF15 is involved in the development of atherosclerosis by regulating apoptotic cell death and IL-6-dependent inflammatory response (Bonaterra et al., 2012). Another study on LDL-deficient mice has demonstrated that leukocyte-specific KO of GDF15 has beneficial effects against atherosclerosis by reducing macrophage plaque infiltration and decreasing necrotic core formation, suggesting a pro-inflammatory role of GDF15 in atherosclerosis (de Jager et al., 2011). In addition, other studies have identified GDF15 as a candidate biomarker of cellular senescence. In particular, GDF15 has been indicated as a component of the senescence-associated secretory phenotype (SASP) (Guo et al., 2019, Basisty et al., 2020, Di Micco et al., 2021), the pro-inflammatory secretome consisting of numerous cytokines, chemokines, growth factors and proteases released from senescent cells into the tissue microenvironment. Guo et al. have demonstrated that senescent fibroblasts secrete GDF15 as a SASP factor essential to promote cell proliferation, migration and invasion in colon adenoma and colorectal cancer cell lines via the MAPK and PI3K signaling pathways (Guo et al., 2019), suggesting that GDF15 may play an important role in promoting cancer progression. Moreover, several studies have found a strong link between GDF15 and presence of cancer. Circulating levels of GDF15 are in fact significantly higher in patients with different types of cancer compared to healthy controls (Kluger et al., 2011, Wang et al., 2014, Wang et al., 2014, Vocka et al., 2018, O'Neill et al., 2020), thus suggesting that GDF15 can be considered as a marker to identify malignancies and a potential therapeutic target for the treatment of different types of cancer. In agreement, a very recent study demonstrated that circulating levels of GDF15 increase with tumor progression and are very high in patients with hepatocellular carcinoma (Myojin et al., 2021). Moreover, GDF15 KO in hepatic stellate cells suppresses liver tumor formation in mice (Myojin et al., 2021).

In the light of these contrasting findings, the understanding of the biological role of GDF15 appears much more complex than expected, especially in the aging process, where a number of stresses able to induce its expression can occur. Therefore, the analysis of its activities cannot be simple and straight-forward, but rather it has always to consider the biological context in which GDF15 acts. In particular, as it has been established that GDF15 is a stress-response molecule, the levels and nature of stress(es) that accompany GDF15 production should be considered. It is possible that, according to the type and duration of stress, a different role for this protein can be envisaged. In this review we will focus on the role of GDF15 during the aging process and will discuss different aspects of its double-sided nature and propose to consider GDF15 as a representative case of antagonistic pleiotropy. We will also discuss GDF15 in a unifying viewpoint based on the life-history theory, an ecological theory used in the study of aging (Selman et al., 2012, Maklakov and Chapman, 2019) that explains resource allocation during stress response through evolutionary concepts.

Section snippets

Evolutionary history of GDF15

GDF15 orthologues have been annotated in mammals, reptiles, amphibians, bony fish, and birds but there is no clear orthologue observed in the genomes of the other two lineages of craniata, hagfish, lampreys, or lower vertebrates (Lockhart et al., 2020).

Phylogenetic analysis of the GDF15 proteins from different vertebrate species showed three main clusters, with one containing the mammalian species and another containing the other vertebrates. This second cluster also showed differentiation into

GDF15 transcription regulation

The transcriptional regulation of GDF15 is very complex, since its promoter contains different cis- and trans-acting promoter elements. Various stresses and stimuli activate differently GDF15 through a number of transcriptional factors (TFs) and non-coding RNAs that converge to modulate its expression.

The mRNA expression of GDF15 is in part regulated by long non-coding RNAs (lncRNAs) and microRNAs (miRs), two classes of non-coding RNAs of about 200 nucleotides or 22 nucleotides in length,

Genetic variability effects on GDF15 concentrations

Recent data demonstrated that GDF15 protein concentration is also influenced by genetic variability. Folkersen et al. (2020) have found four protein Quantitative Trail Loci (pQTLs) (rs2517481, rs1227734, rs60164552, rs112253475) that significantly affect (p-value < 5E-08) the plasma levels of GDF15 protein in individuals of European ancestry. rs1227734-T and rs112253475-A have a positive effect on the concentration of GDF15 protein, whereas rs2517481-C and rs60164552-C have a negative one (

GDF15 regulation at protein level

In addition to the transcriptional regulation, GDF15 is also regulated at translational and maturation levels. GDF15 is translated into a 308 amino acid protein, including a pro-peptide of 167 amino acids and a mature form of 112 amino acids. GDF15 is at first synthesized as GDF15 precursor (pro-GDF15) that then dimerizes to form the pro-GDF15 dimer. The dimeric form is subsequently cleaved at the furine-like cleavage site, RXXR, forming the mature GFD15 form of 112 amino acids that is secreted

GDF15 receptor(s)

One of the reasons why the role of GDF15 is not yet totally clear is due to the limited knowledge on the receptors by which GDF15 binds to target cells. In 2017, four groups simultaneously identified the GDNF α-like receptor (GFRAL) as the only known GDF15 receptor so far, acting through the REarranged during Transfection (RET) co-receptor. To date, it seems that GFRAL expression is limited to the area postrema (AP) and the nucleus of the solitary tract (NTS), two important hindbrain centers

GDF15 effects

As mentioned, GDF15 has many diverse effects, both at systemic and local level. The discovery of GFRAL receptor in the brain suggested that GDF15 acts centrally to control appetite, body weight and energy balance. This was later confirmed by studies in mice where the ablation of GDF15 led to diet-induced obesity (Tran et al., 2018, Patel et al., 2019), demonstrating the fundamental role of GDF15 in the regulation of energy homeostasis. Moreover, GDF15 acts centrally to cause emesis and nausea,

GDF15 in aging and age-related diseases

According to its definition of mitokine, circulating GDF15 is elevated in animal models with mitochondrial dysfunctions, in patients with mitochondrial diseases, as well as in subjects characterized by mitochondrial impairment. Mitochondrial dysfunction is considered a driver of the aging process across several species, from worms to mammals, however, a mild mitochondrial defect can actually contribute to activate an adaptive and beneficial response in all species. This phenomenon, known as

GDF15 as a mediator of a life history program

As discussed above, GDF15 is emerging as a key player in aging and ARDs, but the interpretation of its biological role is still an open issue. Considering the role of GDF15 in the evolutionary history of our species, as mentioned at the beginning of this review, we have adopted an ecological perspective to gather the huge amount of (sometimes contrasting) data on GDF15. One of the most successful ecological theory used in the study of aging is the life history theory that argues that organisms

Conclusions

GDF15 is emerging not only as an important stress response protein involved in many fundamental biological processes, but also as a key player in the aging process. In fact, it results one of the most up-regulated proteins in old age (Tanaka et al., 2018, Conte et al., 2019, Lehallier et al., 2019), a biomarker of many ARDs and a factor associated with decreased survival at old age (Wiklund et al., 2010, Fujita et al., 2016, Conte et al., 2019, Conte et al., 2020a, Conte et al., 2021). The

CRediT authorship contribution statement

MC, AC, CG, VI, SS: literature search, writing of the text; MC, CG, SS: concept; CF, SS: critical discussion.

Declaration of Competing Interest

The authors declare no conflict of interest.

Acknowledgments

The study was partially supported by the Roberto and Cornelia Pallotti Legacy for Cancer Research to S.S. and Ministry of Science and Higher Education at the Lobachevsky State University of Nizhny Novgorod Agreement No. 075–15-2019–871 to C.F.

References (182)

  • S.J. Huh et al.

    Macrophage inhibitory cytokine-1 regulates melanoma vascular development

    Am. J. Pathol.

    (2010)
  • S. Ikeyama et al.

    Expression of the pro-apoptotic gene gadd153/chop is elevated in liver with aging and sensitizes cells to oxidant injury

    J. Biol. Chem.

    (2003)
  • Y. Iwata et al.

    Treatment with GDF15, a TGFβ superfamily protein, induces protective effect on retinal ganglion cells

    Exp. Eye Res.

    (2021)
  • Y.J. Jin et al.

    Macrophage inhibitory cytokine-1 stimulates proliferation of human umbilical vein endothelial cells by up-regulating cyclins D1 and E through the PI3K/Akt-, ERK-, and JNK-dependent AP-1 and E2F activation signaling pathways

    Cell Signal.

    (2012)
  • H. Johnen et al.

    Increased expression of the TGF-b superfamily cytokine MIC-1/GDF15 protects ApoE(-/-) mice from the development of atherosclerosis

    Cardiovasc. Pathol.

    (2012)
  • S.G. Kang et al.

    Differential roles of GDF15 and FGF21 in systemic metabolic adaptation to the mitochondrial integrated stress response

    iScience

    (2021)
  • S.U. Kang et al.

    Expression of NSAID-activated gene-1 by EGCG in head and neck cancer: involvement of ATM-dependent p53 expression

    J. Nutr. Biochem.

    (2013)
  • K. Kannan et al.

    Profile of gene expression regulated by induced p53: connection to the TGF-beta family

    FEBS Lett.

    (2000)
  • D.H. Kim et al.

    Effect of growth differentiation factor-15 secreted by human umbilical cord blood-derived mesenchymal stem cells on amyloid beta levels in in vitro and in vivo models of Alzheimer’s disease

    Biochem Biophys. Res. Commun.

    (2018)
  • H. Kim et al.

    Growth differentiation factor-15 as a biomarker for sarcopenia in aging humans and mice

    Exp. Gerontol.

    (2020)
  • S. Klaus et al.

    Mitochondrial uncoupling and longevity – a role for mitokines?

    Exp. Gerontol.

    (2020)
  • M. Kleinert et al.

    Exercise increases circulating GDF15 in humans

    Mol. Metab.

    (2018)
  • P.X. Li et al.

    Placental transforming growth factor-beta is a downstream mediator of the growth arrest and apoptotic response of tumor cells to DNA damage and p53 overexpression

    J. Biol. Chem.

    (2000)
  • P. Abulizi et al.

    Growth differentiation factor-15 deficiency augments inflammatory response and exacerbates septic heart and renal injury induced by lipopolysaccharide

    Sci. Rep.

    (2017)
  • A.S. Adler et al.

    Motif module map reveals enforcement of aging by continual NF-kappaB activity

    Genes Dev.

    (2007)
  • D.S. Ahmed et al.

    GDF15/GFRAL pathway as a metabolic signature for cachexia in patients with cancer

    J. Cancer

    (2021)
  • J. Alcazar et al.

    Changes in systemic GDF15 across the adult lifespan and their impact on maximal muscle power: the Copenhagen Sarcopenia Study

    J. Cachexia Sarcopenia Muscle

    (2021)
  • V.J. Baggen et al.

    Prognostic value of N-terminal Pro-B-type natriuretic peptide, troponin-T, and growth-differentiation factor 15 in adult congenital heart disease

    Circulation

    (2017)
  • C.R. Balistreri et al.

    NF-κB pathway activators as potential ageing biomarkers: targets for new therapeutic strategies

    Immun. Ageing

    (2013)
  • N. Basisty et al.

    A proteomic atlas of senescence-associated secretomes for aging biomarker development

    PLOS Biol.

    (2020)
  • H.R. Berthoud

    Homeostatic and non-homeostatic pathways involved in the control of food intake and energy balance

    Obesity

    (2006)
  • S.A. Bloch et al.

    Sustained elevation of circulating growth and differentiation factor-15 and a dynamic imbalance in mediators of muscle homeostasis are associated with the development of acute muscle wasting following cardiac surgery

    Crit. Care Med.

    (2013)
  • S.A. Bloch et al.

    Increased expression of GDF-15 may mediate ICU-acquired weakness by down-regulating muscle microRNAs

    Thorax

    (2015)
  • G.A. Bonaterra et al.

    Growth differentiation factor-15 deficiency inhibits atherosclerosis progression by regulating interleukin-6-dependent inflammatory response to vascular injury

    J. Am. Heart Assoc.

    (2012)
  • M.R. Bootcov et al.

    MIC-1, a novel macrophage inhibitory cytokine, is a divergent member of the TGF-beta superfamily

    Proc. Natl. Acad. Sci. USA

    (1997)
  • A.C. Carlsson et al.

    Growth differentiation factor 15 (GDF-15) is a potential biomarker of both diabetic kidney disease and future cardiovascular events in cohorts of individuals with type 2 diabetes: a proteomics approach

    Ups. J. Med Sci.

    (2020)
  • Y.L. Chai et al.

    Growth differentiation factor-15 and white matter hyperintensities in cognitive impairment and dementia

    Medicine

    (2016)
  • B.V.S.K. Chakravarthi et al.

    miR-34a regulates expression of the stathmin-1 oncoprotein and prostate cancer progression

    Mol. Cancer Res.

    (2018)
  • W. Chatoo et al.

    p53 pro-oxidant activity in the central nervous system: implication in aging and neurodegenerative diseases

    Antioxid. Redox Signal.

    (2011)
  • Y.C. Chen et al.

    LRRTM4 and PCSK5 genetic polymorphisms as markers for cognitive impairment in a hypotensive aging population: a genome-wide association study in Taiwan

    J. Clin. Med.

    (2019)
  • J.C. Cheng et al.

    Wild-type p53 attenuates cancer cell motility by inducing growth differentiation factor-15 expression

    Endocrinology

    (2011)
  • S. Choi et al.

    Proprotein convertases in high-density lipoprotein metabolism

    Biomark. Res.

    (2013)
  • K. Chrysovergis et al.

    NAG-1/GDF-15 prevents obesity by increasing thermogenesis, lipolysis and oxidative metabolism

    Int. J. Obes.

    (2014)
  • H.K. Chung et al.

    GDF15 deficiency exacerbates chronic alcohol- and carbon tetrachloride-induced liver injury

    Sci. Rep.

    (2017)
  • M. Conte et al.

    Human aging and longevity are characterized by high levels of mitokines

    J. Gerontol. A Biol. Sci. Med. Sci.

    (2019)
  • M. Conte et al.

    GDF15 plasma level is inversely associated with level of physical activity and correlates with markers of inflammation and muscle weakness

    Front. Immunol.

    (2020)
  • M. Conte et al.

    Mitochondria, immunosenescence and inflammaging: a role for mitokines?

    Semin. Immunopathol.

    (2020)
  • M. Conte et al.

    Disease-specific plasma levels of mitokines FGF21, GDF15, and Humanin in type II diabetes and Alzheimer's disease in comparison with healthy aging

    Geroscience

    (2021)
  • M. Costa-Mattioli et al.

    The integrated stress response: from mechanism to disease

    Science

    (2020)
  • F. Couture et al.

    PACE4 undergoes an oncogenic alternative splicing switch in cancer

    Cancer Res.

    (2017)
  • Cited by (43)

    View all citing articles on Scopus
    View full text