Back to Journals » Journal of Inflammation Research » Volume 17

Role and Mechanism of Growth Differentiation Factor 15 in Chronic Kidney Disease

Authors Tang Y , Liu T, Sun S, Peng Y , Huang X , Wang S , Zhou Z 

Received 23 November 2023

Accepted for publication 25 April 2024

Published 9 May 2024 Volume 2024:17 Pages 2861—2871

DOI https://doi.org/10.2147/JIR.S451398

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Adam D Bachstetter



Yifang Tang,1,* Tao Liu,2,* Shibo Sun,3 Youbo Peng,1 Xiaoxiao Huang,4 Shuangquan Wang,4 Zhu Zhou1

1Department of Nephrology, the First Affiliated Hospital, Kunming Medical University, Kunming, People’s Republic of China; 2Organ Transplantation Center, the First Affiliated Hospital, Kunming Medical University, Kunming, People’s Republic of China; 3Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, People’s Republic of China; 4Department of Nephrology, Xishuangbanna Dai Autonomous Prefecture People’s Hospital, Xishuangbanna, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Zhu Zhou, Department of Nephrology, The First Affiliated Hospital of Kunming Medical University and Yunnan Province Clinical Research Center for Chronic Kidney Disease, No. 295, Xichang Road, Wuhua District, Kunming, People’s Republic of China, Email [email protected] Shibo Sun, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, People’s Republic of China, Email [email protected]

Abstract: GDF-15 is an essential member of the transforming growth factor-beta superfamily. Its functions mainly involve in tissue injury, inflammation, fibrosis, regulation of appetite and weight, development of tumor, and cardiovascular disease. GDF-15 is involved in various signaling pathways, such as MAPK pathway, PI3K/AKT pathway, STAT3 pathway, RET pathway, and SMAD pathway. In addition, several factors such as p53, ROS, and TNF-α participate the regulation of GDF-15. However, the specific mechanism of these factors regulating GDF-15 is still unclear and more research is needed to explore them. GDF-15 mainly improves the function of kidneys in CKD and plays an important role in the prediction of CKD progression and cardiovascular complications. In addition, the role of GDF-15 in the kidney may be related to the SMAD and MAPK pathways. However, the specific mechanism of these pathways remains unclear. Accordingly, more research on the specific mechanism of GDF-15 affecting kidney disease is needed in the future. In conclusion, GDF-15 may be a therapeutic target for kidney disease.

Keywords: chronic kidney disease, GDF-15, biomarker, inflammation, renal protection

Introduction

Chronic kidney disease (CKD) has garnered global attention due to its prevalence, progressive nature, and irreversibility. The renal function of patients with CKD continues to decline and leads to end-stage renal disease (ESRD) or uremia.1,2 It is estimated that CKD causes over 35.8 million disabilities and 1.2 million deaths annually.3,4 It is reported that systemic inflammation and oxidation play a central role in CKD.5–7 Diabetic kidney disease (DKD) stands as one of the most prevalent causes of CKD. Elevated oxidative stress becomes apparent when vascular or glomerular cells are exposed to high glucose levels.8 Impaired mitochondrial function can contribute to DKD.9 It is elucidated that the escalation of oxidative stress levels during the progression of CKD is a pivotal pathological characteristic. Obesity, as a risk factor for diabetes, frequently manifests in diabetes-related conditions, including DKD.10 Following prolonged obesity, inflammation and oxidative stress levels escalate within the glomeruli, leading to proteinuria.10,11 Inflammation not only accelerates CKD but also triggers complications such as malnutrition, atherosclerosis, coronary artery calcification, heart failure, anemia, and bone diseases, which significantly increase the risk of mortality.12–17 Accordingly, it is crucial to explore the relationship between oxidation and inflammatory factors and CKD to prevent CKD and its complications.

It is well known that there are numerous inflammatory factors which trigger the onset or deterioration of diseases, such as TNF-α, IL-6, IL-1β, chemokines, growth factors, and other regulatory factors.18,19 As an important inflammatory factor, growth differentiation factor 15 (GDF-15) has been the focus of increasing studies.20,21 GDF-15 is a member of transforming growth factor-beta (TGF-β) superfamily and is expressed in various tissues, such as heart, pancreas, kidney, and so on.22–24 Levels of GDF-15 expression increases under the conditions of stress or disease.25 Increasing evidence suggests that GDF-15 is associated with cardiovascular diseases, cancer, and diabetes.25–27 It is demonstrated that levels of GDF-15 are closely related to CKD and risk of cardiovascular complications in CKD.28 Moreover, the increase of GDF-15 in CKD frequently causes inflammation, oxidative stress, fibrosis, and apoptosis, which suggest that GDF-15 play an important role in CKD.29,30 In this paper, we reviewed the role and mechanism of GDF-15 in CKD.

The Structure and Functions of GDF-15

The Structure of GDF-15

GDF15 is an essential member of the TGF-β superfamily, and its gene is located on human chromosome 19 and encodes a relatively large precursor protein with two distinct domains: the precursor domain and the mature domain. Most of the precursor domain is cleavaged in cells, resulting in a biologically active C-terminal fragment which is known as the mature domain. Moreover, the mature domain not only forms a stable dimeric structure that enhances its stability in vitro but also possesses a core region composed of multiple β-folds in its three-dimensional conformation, which present similar functional properties to other members of the TGF-β superfamily. More importantly, GDF-15 undergoes various post-translational modifications such as glycosylation, phosphorylation, and lipid modification, which contribute to its increased stability and specific interactions with other biomolecules.31–34 For instance, GDF-15 binds to its receptors to exert its effects leading to specific cellular responses, such as cellular growth, differentiation, and apoptosis.26,29,35 Furthermore, GDF-15 can also block TGF-β receptors and downstream N-Myc signaling pathways, inhibiting apoptosis and collagen production in primary fibroblasts, thereby exerting anti-fibrotic effects on the kidneys.36

The Functions of GDF-15

GDF-15 is a cytokine that exhibits diverse biological functions (Figure 1). It is demonstrated that the expression of GDF-15 increases under the conditions of tissue injury, inflammation, and other pathological conditions to improve damaged tissues and further harm.37–39 It is reported that the GDF-15 inhibits cell apoptosis and consequently supports cell survival under conditions of ischemia, hypoxia, or toxic-induced damage.25,32,35 In addition, GDF-15 inhibits inflammation by reducing the infiltration of inflammatory cells, diminishing the secretion of cytokines and chemokines, and attenuating macrophage and T cell activity to suppress the release of TNF-α, IL-6, and IL-1β. Moreover, GDF-15 facilitates the transformation from these inflammatory cells to anti-inflammatory counterparts, which ultimately reduces the inflammatory response.35,40–43 Furthermore, GDF-15 plays a pivotal role in regulating appetite and weight. It is reported that increase of GDF-15 triggers a decrease in appetite by GDF-15 binding to its receptors of the hypothalamus, resulting in reduced food intake, which impacts body weight.44,45 Authors suggested that GDF-15 in the blood of obese patients presented higher concentrations, which means that a feedback regulatory mechanism responds to excessive energy intake and fat storage in GDF-15 regulating appetite and weight.27,41 Moreover, GDF-15 may contribute to the obesity-related diseases, such as diabetes and cardiovascular disease.26,46,47 Interestingly, GDF-15 demonstrates contradictory dual roles in tumor biology. On the one hand, the upregulation of GDF-15 following stress, inflammation, and tissue injury exerts a pro-apoptotic effect, countering the proliferation and survival of malignant cells.35,48 On the other hand, GDF-15 regulates the tumor microenvironment and promotes the proliferation, angiogenesis, and metastasis of tumor cells in the progression of cancers, such as breast cancer, stomach cancer, and colorectal cancer.40,49–52 In addition, studies suggest that GDF-15 increases rapidly with the occurrence of myocardial ischemia or reperfusion injury to reduce myocardial injury, inhibit inflammatory mediators such as TNF-α and IL-1β, and combat atherosclerosis and myocardial infarction.53,54 GDF-15 regulates smooth muscle cells and inhibits vascular remodeling to maintain vascular health, especially in hypertensive diseases.55,56 Moreover, GDF-15 expression is associated with reduced cardiac function, which may also represent a protective mechanism in vivo.57

Figure 1 The functions of GDF-15.

Abbreviation: GDF-15, growth differentiation factor 15.

GDF-15 and Signaling Pathways

GDF-15 binds to its receptor to trigger a series of signal cascades inside the cell, which cause a profound impact on key processes such as cell growth, migration, survival, and apoptosis. There are several signaling pathways that GDF-15 is involved in (Figure 2).

Figure 2 GDF-15 and signaling pathways.

Abbreviations: GDF-15, growth differentiation factor 15; JAK, janus kinase; STAT3, signal transduction and transcription-activating protein 3; RET, rearranged during transfection; MEK, mitogen-activated protein kinase kinase; ERK, extracellular regulated protein kinases; PIP3, phosphoinosine-triphosphate; AKT, protein kinase B; mTOR, mammalian target of rapamycin; PI3K, phosphoinositide3-kinase; PIP2, phosphoinosine-diphosphate; ROS, reactive oxygen species; TNF-α, tumor necrosis factor α; SMAD, drosophila mothers against decapentaplegic; ALK 1–7, activin-like kinase type 1 receptors.

MAPK Pathway

The mitogen-activated protein kinase (MAPK) pathway is one of the key signal transduction pathways in cells and is widely involved in many physiological and pathological processes such as cell growth, differentiation, migration, apoptosis, and stress response. It is reported that GDF-15 activates MAPK pathways.58,59 GDF-15 activates ERK1/2, a core component of the MAPK family, by binding to its receptor to regulate various upstream kinases, such as Ras, Raf, and MEK. Activation of ERK1/2 further regulates the phosphorylation of a variety of downstream target proteins, thereby affecting cell growth, differentiation, and survival.25,60,61 GDF-15 may exert its specific effects by modulating specific branches or regulatory points of the MAPK pathway. For example, GDF-15 may promote cell survival and proliferation by enhancing the sustained activation of ERK1/2 in vascular smooth muscle cells, while GDF-15 may regulate cellular stress response and apoptosis by regulating JNK or p38 MAPK in HaCaT cells.25,62,63

PI3K/AKT Pathway

The phosphoinositide3-kinase/protein kinase B (PI3K/AKT) pathway plays a central role in cell growth, metabolism, survival, and apoptosis.64–67 PI3K is the primary promoter of PI3K/AKT pathway and is activated by various stimuli such as growth factors and insulin. Activated PI3K converts phosphoinosine-diphosphate (PIP2) to phosphoinosine-triphosphate (PIP3). As a powerful secondary messenger, PIP3 activates AKT which is also known as protein kinase B. Activated AKT subsequently inhibits the function of the pro-apoptotic protein Bcl-2 family, thereby inhibiting apoptosis.68 In addition, AKT activates mammalian target of rapamycin (mTOR) protein to stimulate cell growth and protein synthesis.69 It is reported that the GDF-15 is an important activator of the PI3K/AKT pathway. GDF-15 increases the activity of PI3K to activate AKT in certain stressful environments and pathological states, which is closely related to cell survival and proliferation. It is suggested that GDF-15 significantly reduces cardiomyocyte apoptosis in a myocardial ischemia/reperfusion injury by activating the PI3K/AKT pathway to protect the effect of the heart.70,71 In addition, GDF-15 promotes the survival, migration, and invasion of cancer cells by activating the PI3K/AKT pathway.25,35

STAT3 Pathway

The signal transduction and transcription-activating protein 3 (STAT3) pathway plays a central role in cell growth, differentiation, survival, and immune response and abnormal activation of STAT3 is implicated in various cancers, immune diseases, and inflammation-related diseases.72,73 Biologically, STAT3 is activated by a variety of signals such as growth factors, cytokines, and hormones. These stimuli first bind to their receptors to activate the janus kinase (JAK). Subsequently, JAK phosphorylates STAT3, causing it to form a dimer and transfer to the nucleus, which in turn regulates the target genes.74,75 It is reported that GDF-15 promotes the phosphorylation of STAT3 in the context of inflammation and tumor, thereby affecting cell survival, proliferation, and differentiation.76 Moreover, GDF-15 increases the survival and proliferation ability of breast tumor cells and promotes tumor progression by activating STAT3 pathway.77 In addition, the GDF-15 actives the STAT3 pathway to inhibit inflammation. It is suggested that GDF-15 inhibits the production of inflammatory factors by regulating STAT3 activity in the recruitment of inflammatory cells after myocardial infarction, which present anti-inflammatory effect of GDF-15.78

RET Pathway

Rearranged during transfection (RET) is a transmembrane receptor tyrosine kinase, which is mainly involved in the regulation of neuron growth, differentiation, and migration.79,80 GDF-15 binds to RET co-receptor to promote the activation and self-phosphorylation of RET. Activated RET further activates a variety of downstream signal molecules or pathways, such as Ras/MAPK, PI3K/AKT, or PLCγ pathways to mediate biological effects.81,82 It is reported that high expression of GDF-15 enhances RET activation in tumor cells, thereby promoting tumor cell proliferation and survival.81 In addition, GDF-15 activating RET pathway enhances cell migration and invasion in a variety of tumor models.83 Accordingly, GDF-15 is involved in the RET pathway to regulate cell growth, survival, migration, and invasion.

SMAD Pathway

Drosophila mothers against decapentaplegic (SMAD) proteins constitute a crucial class of signaling transducers, participating in the regulation of various biological processes such as cell proliferation, differentiation, and growth. The SMAD signaling pathway primarily involves transforming growth factor-β (TGF-β), bone morphogenetic protein (BMP), and activin-like kinase (ALK). It has been reported that in pressure-induced cardiac hypertrophy models, GDF-15 inhibits myocardial hypertrophy via the SMAD2/3 pathway.84 Similarly, it also reported that GDF-15 protects the heart from adrenergic-induced hypertrophy through a SMAD-independent pathway.84 GDF-15 exerts cardioprotective effects by activating ALK type 1 receptors (ALK 1–7) and phosphorylating SMAD2/3 and SMAD1/5/8. Phosphorylated SMAD proteins form heteromeric complexes with SMAD4, which are then translocated to the nucleus, activating anti-hypertrophic and anti-apoptotic pathways.30

Other Factors

Although the expression and secretion of GDF-15 is regulated by a variety of physiological and pathological conditions, the exact upstream regulatory factors and mechanisms are still being investigated. It is known that p53, ROS, and TNF-α up-regulate the expression of GDF-15.

As a well-known tumor suppressor protein, p53 is activated and stabilized with the damage of DNA. It is reported that activated p53 directly enhances the transcriptional activity of GDF-15 gene, resulting in an increase of GDF-15.85,86

Reactive oxygen species (ROS) plays a vital role in cells and is involved in many important biological processes, such as signal transduction, gene expression regulation, and cell growth.87,88 Excess ROS frequently leads to cell damage, leading to a variety of diseases, such as cancer, cardiovascular disease, neurodegenerative diseases, and premature aging.89–91 It is presented that high ROS induced by oxidative stress significantly elevates the expression and accumulation of GDF-15.92 Moreover, the upregulation of GDF-15 modulates antioxidant expression or activates antioxidant pathways to reduce the cell damage caused by ROS.48 However, persistent GDF-15 overexpression often means continuous oxidative stress, which contributes to cellular dysfunction and disease progression.

Tumor necrosis factor α (TNF-α) is a multifunctional cytokine, which plays a key role in inflammation, immune response, cell proliferation, and apoptosis.93,94 TNF-α binding to its receptor activates multiple signal transduction pathways, especially the classical NF-κB and MAPK pathways, thereby enhancing the transcriptional activity of GDF-15.30 In addition, TNF-α affects the stability of GDF-15 mRNA or interacts with microRNAs, such as miR-20a and miR-221.95,96

The Role of GDF-15 in the Kidney Diseases

Renal Protection Provided by GDF-15

The role of GDF-15 presents potential renal protective effects in kidney disease and injury, especially during inflammation, injury, and recovery.97 It is shown that the loss of GDF-15 enhances the inflammatory response, which is harmful for lipopolysaccharide-induced (LPS-induced) kidney and heart damage, while overexpression of GDF-15 improves LPS-induced organ dysfunction.98 In addition, GDF-15 is related to early renal protective injury responses by altering the behavior of immune cells, such as T cells.81 It is reported that the mice deficient in GDF-15 exhibited more severe symptoms, including increased proteinuria, crescent formation, and mesangial dilation in a model of anti-glomerular basement membrane glomerulonephritis,35 which means the critical role of GDF-15 in regulating T cells, particularly their chemotaxis within the kidney.99 In addition, GDF-15 protects the renal interstitial and tubule compartments, and deletion or absence of this GDF-15 gene results in increased damage of tubule and interstitial in diabetes, though the glomerular damage is not impaired.26,100 Moreover, renal injury causes a notable upregulation of GDF-15 at the proximal renal tubule site, which induces up-expression of Klotho protein to improve kidney injury.101 Accordingly, GDF-15 presents the renal protection.

Chronic Kidney Disease

CKD is a global health issue due to its association with multiple complications and adverse outcomes.102 In recent years, the research mainly focuses on the predictive value of GDF-15 for the progression of CKD, especially for diabetic nephropathy. In addition, another focus is the role of GDF-15 in the occurrence of complications of CKD, such as cardiovascular risk, renal fibrosis, mineral bone disease, and anemia.

Meanwhile, elevation of GDF-15 is related to adverse outcomes of kidney disease, which suggest GDF-15 may be an early warning biomarker for CKD.103 It is reported that circulating levels of GDF-15 are correlated with renal expression of GDF-15, which is significantly associated with the risk of CKD progression,29 which suggests that circulating GDF-15 can independently predict CKD progression and poor prognosis.98 Moreover, high levels of GDF-15 are significantly associated with increased all-cause and cardiovascular mortality and morbidity in patients with diabetic kidney disease. Notably, this association appears to reflect a more rapid deterioration of renal function than being directly related to the development of ESRD.104 Accordingly, GDF-15 may play an important role in early prediction of kidney function decline. In addition, it is confirmed that high circulating levels of GDF-15 are associated with type 2 diabetic nephropathy, which means GDF-15 may be a potential biomarker for type 2 diabetic nephropathy.105 It is reported that the GDF-15 is an important activator of the PI3K/AKT pathway. The PI3K/AKT signaling pathway attenuates cellular apoptosis in CKD. Glucagon-like peptide-1 (GLP-1) receptor agonists, commonly utilized as fundamental therapeutic agents in CKD,106 inhibit protein kinase C (PKC)β activation,107 promote insulin/IRS1 signaling, and via the PI3K/Akt pathway, mediate increased production of nitric oxide (NO), inducing vasodilation and inhibiting podocyte apoptosis.108 Consequently, GDF-15 can suppress CKD cellular apoptosis via the PI3K/AKT signaling pathway, exerting renal protective effects.

GDF-15 is also involved in the occurrence of complications in CKD.109–111 It is reported that GDF-15 is independently associated with cardiovascular risk in patients with diabetic nephropathy.104 GDF-15 is more closely associated with microvascular complications, especially diabetic nephropathy, than with macrovascular complications.81 In addition, GDF-15 is more closely associated with heart failure, especially heart failure with preserved ejection fraction in CKD.112,113 Moreover, GDF-15 is related with the complications such as renal fibrosis.29 It is suggested that GDF-15 plays a role in the development of mineral bone disorders due to its association with vitamin D synthesis and phosphorus metabolism.114 Additionally, elevated levels of GDF-15 are correlated with reduced erythropoiesis and impaired iron utilization, which may lead to anemia in CKD.115,116

Kidney Transplantation

GDF-15 plays an important role in post-transplant outcomes. The prevalence of anemia among kidney transplant recipients is notably high, primarily attributed to long-term uremia or the nephrotoxicity of immunosuppressive drugs.117 It is suggested that there is a plausible link between GDF-15 and anemia in patients with kidney transplantation. Intriguingly, GDF-15 levels reduce in kidney transplantation, while increases in single nephrectomy.118 It seems that elevated levels of GDF-15 may reflect the presence of chronic kidney disease rather than being solely attributable to transplantation itself. Notably, there is an intriguing correlation between GDF-15 and mortality following kidney transplantation,119 which suggest that GDF-15 may be a predictive indicator for early mortality after surgery. Remarkably, this predictive power even enhances the accuracy of the EPTS score which is a scoring model used to assess expected post-transplant survival among candidates for kidney transplantation.119 Apart from its prognostic value regarding mortality rates, GDF-15 also serves as a key predictor for cardiovascular events.118

Conclusions and Prospects

GDF-15 plays a crucial role in tissue injury, inflammation, fibrosis, regulation of appetite and weight, tumor development, cardiovascular disease, and other biological functions following the processes of glycosylation and phosphorylation. GDF-15 is involved in various signaling pathways such as MAPK pathway, PI3K/AKT pathway, STAT3 pathway, RET pathway, and SMAD pathway. In addition, GDF-15 is regulated by several factors such as p53, ROS, and TNF-α. In CKD, GDF-15 are strongly associated with CKD survival rate, disease progression rate, cardiovascular complications risk, and complications such as renal fibrosis and anemia. Particularly, GDF-15 are linked to both decreased kidney function and higher risk for cardiovascular diseases in diabetic nephropathy patients. Moreover, GDF-15 has shown potential value in predicting post-transplantation outcomes, such as anemia occurrence, cardiovascular events, and mortality rates. However, the specific mechanism that GDF-15 affects kidneys remains unclear. In addition, more research is needed to explore the role of GDF-15 in common CKD, such as IgA nephropathy, membranous nephropathy, and lupus nephritis. In conclusion, GDF-15 may be a therapeutic target.

Funding

This work was supported by Yunnan Major Scientific and Technological Project, (grant no. 202102AA100060), Yunnan Revitalization Talent Support Program, the National Natural Science Foundation of China (grant no. 82160007), the Yunnan Provincial Science and Technology Department (grant no. 2019FE001(−58), 202201AY070001(−074)), Yunnan Province young and middle-aged academic and technical leaders reserve talent project (grant no. 202305AC160017), 535 Talent Project of First Affiliated Hospital of Kunming Medical University (grant no. 2023535D12), and Student Innovation Fund project of Kunming Medical University (grant no.2022JXD212, 2022JXD227).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Kovesdy CP. Epidemiology of chronic kidney disease: an update 2022. Kidney Int Suppl. 2022;12(1):7–11. doi:10.1016/j.kisu.2021.11.003

2. Kalantar-Zadeh K, Jafar TH, Nitsch D, Neuen BL, Perkovic V. Chronic kidney disease. Lancet. 2021;398:786–802.

3. Mrug M, Bloom MS, Seto C, et al. Genetic testing for chronic kidney diseases: clinical utility and barriers perceived by nephrologists. Kidney Med. 2021;3(6):1050–1056. doi:10.1016/j.xkme.2021.08.006

4. Jdiaa SS, Mansour R, El Alayli A, Gautam A, Thomas P, Mustafa RA. COVID-19 and chronic kidney disease: an updated overview of reviews. J Nephrol. 2022;35(1):69–85. doi:10.1007/s40620-021-01206-8

5. Bikbov B, Purcell CA, Levey AS. Global, regional, and national burden of chronic kidney disease, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2020;395(10225):709–733. doi:10.1016/S0140-6736(20)30045-3

6. Irazabal MV, Torres VE. Reactive oxygen species and redox signaling in chronic kidney disease. Cells. 2020;9(6):1342. doi:10.3390/cells9061342

7. Tirichen H, Yaigoub H, Xu W, Wu C, Li R, Li Y. Mitochondrial reactive oxygen species and their contribution in chronic kidney disease progression through oxidative stress. Front Physiol. 2021;12:627837. doi:10.3389/fphys.2021.627837

8. Inoguchi T, Sonta T, Tsubouchi H, et al. Protein kinase C-dependent increase in reactive oxygen species (ROS) production in vascular tissues of diabetes: role of vascular NAD(P)H oxidase. J Am Soc Nephrol. 2003;14(suppl_3):S227–232. doi:10.1097/01.ASN.0000077407.90309.65

9. Mima A. Mitochondria-targeted drugs for diabetic kidney disease. Heliyon. 2022;8(2):e08878. doi:10.1016/j.heliyon.2022.e08878

10. Mima A. Inflammation and oxidative stress in diabetic nephropathy: new insights on its inhibition as new therapeutic targets. J Diabetes Res. 2013;2013:248563. doi:10.1155/2013/248563

11. Mima A, Yasuzawa T, King GL, Ueshima S. Obesity-associated glomerular inflammation increases albuminuria without renal histological changes. FEBS Open Bio. 2018;8(4):664–670. doi:10.1002/2211-5463.12400

12. Graterol Torres F, Molina M, Soler-Majoral J, et al. Evolving concepts on inflammatory biomarkers and malnutrition in chronic kidney disease. Nutrients. 2022;15(1):14. doi:10.3390/nu15010014

13. Valdivielso JM, Rodríguez-Puyol D, Pascual J, et al. Atherosclerosis in chronic kidney disease: more, less, or just different? Arterioscler. Thromb Vasc Biol. 2019;39(10):1938–1966. doi:10.1161/ATVBAHA.119.312705

14. Wang XR, Zhang JJ, Xu XX, Wu YG. Prevalence of coronary artery calcification and its association with mortality, cardiovascular events in patients with chronic kidney disease: a systematic review and meta-analysis. Ren Fail. 2019;41(1):244–256. doi:10.1080/0886022X.2019.1595646

15. van de Wouw J, Broekhuizen M, Sorop O, et al. Chronic kidney disease as a risk factor for heart failure with preserved ejection fraction: a focus on microcirculatory factors and therapeutic targets. Front Physiol. 2019;10:1108. doi:10.3389/fphys.2019.01108

16. Hanna RM, Streja E, Kalantar-Zadeh K. Burden of anemia in chronic kidney disease: beyond erythropoietin. Adv Ther. 2021;38(1):52–75. doi:10.1007/s12325-020-01524-6

17. Cannata-Andía JB, Martín-Carro B, Martín-Vírgala J, et al. Chronic kidney disease-mineral and bone disorders: pathogenesis and management. Calcif Tissue Int. 2021;108(4):410–422. doi:10.1007/s00223-020-00777-1

18. Ghassib I, Chen Z, Zhu J, Wang HL. Use of IL-1 β, IL-6, TNF-α, and MMP-8 biomarkers to distinguish peri-implant diseases: a systematic review and meta-analysis. Clin Implant Dent Relat Res. 2019;21(1):190–207. doi:10.1111/cid.12694

19. Hammad AM, Ibrahim YA, Khdair SI, et al. Metformin reduces oxandrolone- induced depression-like behavior in rats via modulating the expression of IL-1β, IL-6, IL-10 and TNF-α. Behav Brain Res. 2021;414:113475. doi:10.1016/j.bbr.2021.113475

20. Bermúdez B, López S, Pacheco YM, et al. Influence of postprandial triglyceride-rich lipoproteins on lipid-mediated gene expression in smooth muscle cells of the human coronary artery. Cardiovasc Res. 2008;79(2):294–303. doi:10.1093/cvr/cvn082

21. Ding Q, Mracek T, Gonzalez-Muniesa P, et al. Identification of macrophage inhibitory cytokine-1 in adipose tissue and its secretion as an adipokine by human adipocytes. Endocrinology. 2009;150(4):1688–1696. doi:10.1210/en.2008-0952

22. Yokoyama-Kobayashi M, Saeki M, Sekine S, Kato S. Human cDNA encoding a novel TGF-beta superfamily protein highly expressed in placenta. J Biochem. 1997;122(3):622–626. doi:10.1093/oxfordjournals.jbchem.a021798

23. Koopmann J, Buckhaults P, Brown DA, et al. Serum macrophage inhibitory cytokine 1 as a marker of pancreatic and other periampullary cancers. Clin Cancer Res. 2004;10(7):2386–2392. doi:10.1158/1078-0432.CCR-03-0165

24. Tan M, Wang Y, Guan K, Sun Y. PTGF-β, a type β transforming growth factor (TGF-β) superfamily member, is a p53 target gene that inhibits tumor cell growth via TGF-β signaling pathway. Proc Natl Acad Sci U S A. 2000;97(1):109–114. doi:10.1073/pnas.97.1.109

25. Fang L, Li F, Gu C. GDF-15: a multifunctional modulator and potential therapeutic target in cancer. Curr Pharm Des. 2019;25(6):654–662. doi:10.2174/1381612825666190402101143

26. Xiao QA, He Q, Zeng J, Xia X. GDF-15, a future therapeutic target of glucolipid metabolic disorders and cardiovascular disease. Biomed Pharmacother. 2022;146:112582. doi:10.1016/j.biopha.2021.112582

27. Ouyang J, Isnard S, Lin J, et al. GDF-15 as a weight watcher for diabetic and non-diabetic people treated with metformin. Front Endocrinol. 2020;11:581839. doi:10.3389/fendo.2020.581839

28. Benes J, Kotrc M, Wohlfahrt P, et al. The Role of GDF-15 in heart failure patients with chronic kidney disease. Can J Cardiol. 2019;35(4):462–470. doi:10.1016/j.cjca.2018.12.027

29. Tuegel C, Katz R, Alam M, et al. GDF-15, galectin 3, soluble ST2, and risk of mortality and cardiovascular events in CKD. Am J Kidney Dis. 2018;72(4):519–528. doi:10.1053/j.ajkd.2018.03.025

30. Adela R, Banerjee SK. GDF-15 as a target and biomarker for diabetes and cardiovascular diseases: a translational prospective. J Diabetes Res. 2015;2015:490842. doi:10.1155/2015/490842

31. Zhao Z, Zhang J, Yin L, et al. Upregulated GDF-15 expression facilitates pancreatic ductal adenocarcinoma progression through orphan receptor GFRAL. Aging. 2020;12(22):22564–22581. doi:10.18632/aging.103830

32. Verhamme FM, Freeman CM, Brusselle GG, Bracke KR, Curtis JL. GDF-15 in pulmonary and critical care medicine. Am J Respir Cell Mol Biol. 2019;60(6):621–628. doi:10.1165/rcmb.2018-0379TR

33. Desmedt S, Desmedt V, De Vos L, Delanghe JR, Speeckaert R, Speeckaert MM. Growth differentiation factor 15: a novel biomarker with high clinical potential. Crit Rev Clin Lab Sci. 2019;56(5):333–350. doi:10.1080/10408363.2019.1615034

34. Sithiravel C, Røysland R, Alaour B, et al. Biological variation, reference change values and index of individuality of GDF-15. Clin Chem Lab Med. 2022;60(4):593–596. doi:10.1515/cclm-2021-0769

35. Wischhusen J, Melero I, Fridman WH. Growth/differentiation factor-15 (GDF-15): from biomarker to novel targetable immune checkpoint. Front Immunol. 2020;11:951. doi:10.3389/fimmu.2020.00951

36. Kim YI, Shin HW, Chun YS, Park JW. CST3 and GDF15 ameliorate renal fibrosis by inhibiting fibroblast growth and activation. Biochem Biophys Res Commun. 2018;500(2):288–295. doi:10.1016/j.bbrc.2018.04.061

37. Ahmed DS, Isnard S, Berini C, Lin J, Routy JP, Royston L. Coping with stress: the mitokine GDF-15 as a biomarker of COVID-19 severity. Front Immunol. 2022;13:820350. doi:10.3389/fimmu.2022.820350

38. Mastrobattista E, Lenze EJ, Reynolds CF, et al. Late-life depression is associated with increased levels of GDF-15, a pro-aging mitokine. Am J Geriatr Psychiatry. 2023;31(1):1–9. doi:10.1016/j.jagp.2022.08.003

39. Yang M, Darwish T, Larraufie P, et al. Inhibition of mitochondrial function by metformin increases glucose uptake, glycolysis and GDF-15 release from intestinal cells. Sci Rep. 2021;11(1):2529. doi:10.1038/s41598-021-81349-7

40. Lodi RS, Yu B, Xia L, Liu F. Roles and regulation of growth differentiation factor-15 in the immune and tumor microenvironment. Hum Immunol. 2021;82(12):937–944. doi:10.1016/j.humimm.2021.06.007

41. Schwarz A, Kinscherf R, Bonaterra GA. Role of the stress- and inflammation-induced cytokine GDF-15 in cardiovascular diseases: from basic research to clinical relevance. RCM. 2023;2:24.

42. Roth P, Junker M, Tritschler I, et al. GDF-15 contributes to proliferation and immune escape of malignant gliomas. Clin Cancer Res. 2010;16(15):3851–3859. doi:10.1158/1078-0432.CCR-10-0705

43. Chen J, Luo F, Fang Z, Zhang W. GDF-15 levels and atherosclerosis. Int J Cardiol. 2018;257:36. doi:10.1016/j.ijcard.2017.10.037

44. Blanco AM, Bertucci JI, Velasco C, Unniappan S. Growth differentiation factor 15 (GDF-15) is a novel orexigen in fish. Mol Cell Endocrinol. 2020;505:110720. doi:10.1016/j.mce.2020.110720

45. Molfino A, Amabile MI, Imbimbo G, et al. Association between growth differentiation factor-15 (GDF-15) serum levels, anorexia and low muscle mass among cancer patients. Cancers. 2020;13(1):13. doi:10.3390/cancers13010013

46. Valenzuela-Vallejo L, Chrysafi P, Bello-Ramos J, Bsata S, Mantzoros CS. Circulating total and intact GDF-15 levels are not altered in response to weight loss induced by liraglutide or lorcaserin treatment in humans with obesity. Metabolism. 2022;133:155237. doi:10.1016/j.metabol.2022.155237

47. Merchant RA, Chan YH, Duque G, Bencivenga L. GDF-15 is associated with poor physical function in prefrail older adults with diabetes. J Diabetes Res. 2023;2023:2519128. doi:10.1155/2023/2519128

48. Verhamme FM, Seys LJM, De Smet EG, et al. Elevated GDF-15 contributes to pulmonary inflammation upon cigarette smoke exposure. Mucosal Immunol. 2017;10(6):1400–1411. doi:10.1038/mi.2017.3

49. Mielcarska S, Stopińska K, Dawidowicz M, et al. GDF-15 level correlates with CMKLR1 and VEGF-A in tumor-free margin in colorectal cancer. Curr Med Sci. 2021;41(3):522–528. doi:10.1007/s11596-021-2335-0

50. Hasanpour Segherlou Z, Nouri-Vaskeh M, Noroozi Guilandehi S, et al. GDF-15: diagnostic, prognostic, and therapeutic significance in glioblastoma multiforme. J Cell Physiol. 2021;236(8):5564–5581. doi:10.1002/jcp.30289

51. Zhang H, Zhou Y, Cui B, Liu Z, Shen H. Novel insights into astrocyte-mediated signaling of proliferation, invasion and tumor immune microenvironment in glioblastoma. Biomed Pharmacother. 2020;126:110086. doi:10.1016/j.biopha.2020.110086

52. Haake M, Haack B, Schäfer T, et al. Tumor-derived GDF-15 blocks LFA-1 dependent T cell recruitment and suppresses responses to anti-PD-1 treatment. Nat Commun. 2023;14(1):4253. doi:10.1038/s41467-023-39817-3

53. Preusch MR, Baeuerle M, Albrecht C, et al. GDF-15 protects from macrophage accumulation in a mousemodel of advanced atherosclerosis. Eur J Med Res. 2013;18(1):19. doi:10.1186/2047-783X-18-19

54. Maimaiti Y, Cheng H, Guo Z, Yu X, Tuohuti A, Li G. Correlation between serum GDF-15 level and pulmonary vascular morphological changes and prognosis in patients with pulmonary arterial hypertension. Front Cardiovasc Med. 2023;10:1085122. doi:10.3389/fcvm.2023.1085122

55. May BM, Pimentel M, Zimerman LI, Rohde LE. GDF-15 as a Biomarker in Cardiovascular Disease. Arq Bras Cardiol. 2021;116(3):494–500. doi:10.36660/abc.20200426

56. Bonaterra GA, Struck N, Zuegel S, et al. Characterization of atherosclerotic plaques in blood vessels with low oxygenated blood and blood pressure (Pulmonary trunk): role of growth differentiation factor-15 (GDF-15). BMC Cardiovasc Disord. 2021;21(1):601. doi:10.1186/s12872-021-02420-9

57. Bettencourt P, Ferreira-Coimbra J, Rodrigues P, et al. Towards a multi-marker prognostic strategy in acute heart failure: a role for GDF-15. ESC Heart Fail. 2018;5(6):1017–1022. doi:10.1002/ehf2.12301

58. Braicu C, Buse M, Busuioc C, et al. A comprehensive review on MAPK: a promising therapeutic target in cancer. Cancers. 2019;12(1):11. doi:10.3390/cancers12010011

59. Lee S, Rauch J, Kolch W. Targeting MAPK signaling in cancer: mechanisms of drug resistance and sensitivity. Int J Mol Sci. 2020;17(1):21. doi:10.7150/ijms.39074

60. Sun Y, Lin J, Zhao T, Zhang L. GDF-15 promotes proliferation of vascular smooth muscle cells through MAPK-activated protein kinase pathways. Int J Clin Exp Med. 2019;12:10093–10100.

61. Wang SF, Chen S, Tseng LM, Lee HC. Role of the mitochondrial stress response in human cancer progression. Exp Biol Med. 2020;245(10):861–878. doi:10.1177/1535370220920558

62. Dong G, Zheng QD, Ma M, et al. Angiogenesis enhanced by treatment damage to hepatocellular carcinoma through the release of GDF15. Cancer Med. 2018;7(3):820–830. doi:10.1002/cam4.1330

63. Decean HP, Brie IC, Tatomir CB, Perde-Schrepler M, Fischer-Fodor E, Virag P. Targeting MAPK (p38, ERK, JNK) and inflammatory CK (GDF-15, GM-CSF) in UVB-activated human skin cells with Vitis vinifera seed extract. J Environ Pathol Toxicol Oncol. 2018;37(3):261–272. doi:10.1615/JEnvironPatholToxicolOncol.2018027009

64. Jafari M, Ghadami E, Dadkhah T, Akhavan-Niaki H. PI3k/AKT signaling pathway: erythropoiesis and beyond. J Cell Physiol. 2019;234(3):2373–2385. doi:10.1002/jcp.27262

65. Very N, Vercoutter-Edouart AS, Lefebvre T, Hardivillé S, El Yazidi-Belkoura I. Cross-dysregulation of O-GlcNAcylation and PI3K/AKT/mTOR axis in human chronic diseases. Front Endocrinol. 2018;9:602. doi:10.3389/fendo.2018.00602

66. Nur Husna SM, Tan HT, Mohamud R, Dyhl-Polk A, Wong KK. Inhibitors targeting CDK4/6, PARP and PI3K in breast cancer: a review. Ther Adv Med Oncol. 2018;10:1758835918808509. doi:10.1177/1758835918808509

67. Matsuda S, Ikeda Y, Murakami M, Nakagawa Y, Tsuji A, Kitagishi Y, Roles of PI3K/AKT/GSK3 Pathway Involved in Psychiatric Illnesses. Diseases. 2019;1:7. doi:10.3390/diseases7010022

68. Li L, Wang F, Zhang J, et al. Typical phthalic acid esters induce apoptosis by regulating the PI3K/Akt/Bcl-2 signaling pathway in rat insulinoma cells. Ecotoxicol Environ Saf. 2021;208:111461. doi:10.1016/j.ecoenv.2020.111461

69. Alzahrani AS. PI3K/Akt/mTOR inhibitors in cancer: at the bench and bedside. Semin Cancer Biol. 2019;59:125–132. doi:10.1016/j.semcancer.2019.07.009

70. Arkoumani M, Papadopoulou-Marketou N, Nicolaides NC, Kanaka-Gantenbein C, Tentolouris N, Papassotiriou I. The clinical impact of growth differentiation factor-15 in heart disease: a 2019 update. Crit Rev Clin Lab Sci. 2020;57(2):114–125. doi:10.1080/10408363.2019.1678565

71. Xiangrui Q, Junhui L, Rui H, Xiaozhen Z. GDF-15 in plasma and circulating mononuclear cells and NT-proBNP for diagnosis of chronic heart failure and predicting cardiovascular disease events. Nan Fang Yi Ke Da Xue Xue Bao. 2019;39:1273–1279. doi:10.12122/j.issn.1673-4254.2019.11.02

72. Zou S, Tong Q, Liu B, Huang W, Tian Y, Fu X. Targeting STAT3 in cancer immunotherapy. Mol Cancer. 2020;19(1):145. doi:10.1186/s12943-020-01258-7

73. Wang HQ, Man QW, Huo FY, et al. STAT3 pathway in cancers: past, present, and future. MedComm. 2022;3(2):e124. doi:10.1002/mco2.124

74. Jin W. Role of JAK/STAT3 signaling in the regulation of metastasis, the transition of cancer stem cells, and chemoresistance of cancer by epithelial-mesenchymal transition. Cells. 2020;9(1):217. doi:10.3390/cells9010217

75. Wang T, Fahrmann JF, Lee H, et al. JAK/STAT3-regulated fatty acid β-oxidation is critical for breast cancer stem cell self-renewal and chemoresistance. Cell Metab. 2018;27(1):136–150.e135. doi:10.1016/j.cmet.2017.11.001

76. Park JY, Yoo KD, Shin SJ, Kim KS, Kim YS, Yang SH. FP260Inhibition of CXCR3 expression through blockade of STAT3 alpha signaling down-regulate inflammation of renal ischemia-reperfusion injury. Nephrol Dial Transplant. 2019;34(Supplement_1). doi:10.1093/ndt/gfz106.FP260

77. Blanchette-Farra N, Kita D, Konstorum A, et al. Contribution of three-dimensional architecture and tumor-associated fibroblasts to hepcidin regulation in breast cancer. Oncogene. 2018;37(29):4013–4032. doi:10.1038/s41388-018-0243-y

78. Kempf T, Zarbock A, Widera C, et al. GDF-15 is an inhibitor of leukocyte integrin activation required for survival after myocardial infarction in mice. Nat Med. 2011;17(5):581–588. doi:10.1038/nm.2354

79. Enomoto H, Crawford PA, Gorodinsky A, Heuckeroth RO, Johnson EM, Milbrandt J. RET signaling is essential for migration, axonal growth and axon guidance of developing sympathetic neurons. Development. 2001;128(20):3963–3974. doi:10.1242/dev.128.20.3963

80. Enomoto H, Heuckeroth RO, Golden JP, Johnson EM, Milbrandt J. Development of cranial parasympathetic ganglia requires sequential actions of GDNF and neurturin. Development. 2000;127(22):4877–4889. doi:10.1242/dev.127.22.4877

81. Iglesias P, Silvestre RA, Díez JJ. Growth differentiation factor 15 (GDF-15) in endocrinology. Endocrine. 2023;81(3):419–431. doi:10.1007/s12020-023-03377-9

82. Bou Antoun N, Chioni AM. Dysregulated signalling pathways driving anticancer drug resistance. Int J Mol Sci. 2023;25(1):24. doi:10.3390/ijms25010024

83. Muniyan S, Pothuraju R, Seshacharyulu P, Batra SK. Macrophage inhibitory cytokine-1 in cancer: beyond the cellular phenotype. Cancer Lett. 2022;536:215664. doi:10.1016/j.canlet.2022.215664

84. Xu J, Kimball TR, Lorenz JN, et al. GDF15/MIC-1 functions as a protective and antihypertrophic factor released from the myocardium in association with SMAD protein activation. Circul Res. 2006;98(3):342–350. doi:10.1161/01.RES.0000202804.84885.d0

85. Heduschke A, Ackermann K, Wilhelm B, et al. GDF-15 deficiency reduces autophagic activity in human macrophages in vitro and decreases p62-accumulation in atherosclerotic lesions in mice. Cells. 2021;11(1):10. doi:10.3390/cells11010010

86. Ranjbaran R, Abbasi M, Rahimian E, et al. GDF-15 negatively regulates excess erythropoiesis and its overexpression is involved in erythroid hyperplasia. Exp Cell Res. 2020;397(2):112346. doi:10.1016/j.yexcr.2020.112346

87. Perillo B, Di Donato M, Pezone A, et al. ROS in cancer therapy: the bright side of the moon. Exp Mol Med. 2020;52(2):192–203. doi:10.1038/s12276-020-0384-2

88. Nakamura H, Takada K. Reactive oxygen species in cancer: current findings and future directions. Cancer Sci. 2021;112:3945–3952.

89. Madreiter-Sokolowski CT, Thomas C, Ristow M. Interrelation between ROS and Ca(2+) in aging and age-related diseases. Redox Biol. 2020;36:101678. doi:10.1016/j.redox.2020.101678

90. Hajam YA, Rani R, Ganie SY, et al. Oxidative stress in human pathology and aging: molecular mechanisms and perspectives. Cells. 2022;12(1):11. doi:10.3390/cells12010011

91. Guo J, Huang X, Dou L, et al. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther. 2022;7(1):391. doi:10.1038/s41392-022-01251-0

92. Ma Y, Zheng L, Wang Y, Gao Y, Xu Y. Arachidonic acid in follicular fluid of PCOS induces oxidative stress in a human ovarian granulosa tumor cell line (KGN) and Upregulates GDF15 expression as a response. Front Endocrinol. 2022;13:865748. doi:10.3389/fendo.2022.865748

93. Wang Y, Che M, Xin J, Zheng Z, Li J, Zhang S. The role of IL-1β and TNF-α in intervertebral disc degeneration. Biomed Pharmacother. 2020;131:110660. doi:10.1016/j.biopha.2020.110660

94. Jang DI, Lee AH, Shin HY, et al. The Role of Tumor Necrosis Factor Alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics. Int J Mol Sci. 2021;23(1):22. doi:10.3390/ijms23010022

95. Piek A, Du W, de Boer RA, Silljé HHW. Novel heart failure biomarkers: why do we fail to exploit their potential? Crit Rev Clin Lab Sci. 2018;55(4):246–263. doi:10.1080/10408363.2018.1460576

96. Zhang BC, Zhang J, Sun L. In-depth profiling and analysis of host and viral microRNAs in Japanese flounder (Paralichthys olivaceus) infected with megalocytivirus reveal involvement of microRNAs in host-virus interaction in teleost fish. BMC Genomics. 2014;15(1):878. doi:10.1186/1471-2164-15-878

97. Delrue C, Speeckaert R, Delanghe JR, Speeckaert MM. Growth differentiation factor 15 (GDF-15) in kidney diseases. Adv Clin Chem. 2023;114:1–46. doi:10.1016/bs.acc.2023.02.003

98. Zhou Z, Liu H, Ju H, Chen H, Jin H, Sun M. Circulating GDF-15 in relation to the progression and prognosis of chronic kidney disease: a systematic review and dose-response meta-analysis. Eur J Int Med. 2023;110:77–85. doi:10.1016/j.ejim.2023.01.026

99. Moschovaki-Filippidou F, Steiger S, Lorenz G, et al. Growth differentiation factor 15 ameliorates anti-glomerular basement membrane glomerulonephritis in mice. Int J Mol Sci. 2020;22(1):21. doi:10.3390/ijms22010021

100. Hamon SM, Griffin TP, Islam MN, Wall D, Griffin MD, O’Shea PM. Defining reference intervals for a serum growth differentiation factor-15 (GDF-15) assay in a Caucasian population and its potential utility in diabetic kidney disease (DKD). Clin Chem Lab Med. 2019;57(4):510–520. doi:10.1515/cclm-2018-0534

101. Kageyama K, Iwasaki Y, Watanuki Y, et al. Growth differentiation factor-15 modulates adrenocorticotropic hormone synthesis in murine AtT-20 corticotroph cells. Peptides. 2022;155:170841. doi:10.1016/j.peptides.2022.170841

102. van Haalen H, Jackson J, Spinowitz B, Milligan G, Moon R. Impact of chronic kidney disease and anemia on health-related quality of life and work productivity: analysis of multinational real-world data. BMC Nephrol. 2020;21(1):88. doi:10.1186/s12882-020-01746-4

103. Perez-Gomez MV, Pizarro-Sanchez S, Gracia-Iguacel C, et al. Urinary Growth Differentiation Factor-15 (GDF15) levels as a biomarker of adverse outcomes and biopsy findings in chronic kidney disease. J Nephrol. 2021;34(6):1819–1832. doi:10.1007/s40620-021-01020-2

104. Carlsson AC, Nowak C, Lind L, 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;125(1):37–43. doi:10.1080/03009734.2019.1696430

105. Barma M, Khan F, Price RJG, et al. Association between GDF-15 levels and changes in vascular and physical function in older patients with hypertension. Aging Clin Exp Res. 2017;29:1055–1059.

106. Mima A, Kitada M, Geraldes P, et al. Glomerular VEGF resistance induced by PKCδ/SHP-1 activation and contribution to diabetic nephropathy. FASEB j. 2012;26(7):2963–2974. doi:10.1096/fj.11-202994

107. Mima A, Nomura A, Fujii T. Current findings on the efficacy of incretin-based drugs for diabetic kidney disease: a narrative review. Biomed Pharmacothe. 2023;165:115032. doi:10.1016/j.biopha.2023.115032

108. Mima A, Yasuzawa T, Nakamura T, Ueshima S. Linagliptin affects IRS1/Akt signaling and prevents high glucose-induced apoptosis in podocytes. Sci Rep. 2020;10(1):5775. doi:10.1038/s41598-020-62579-7

109. Lukaszyk E, Lukaszyk M, Koc-Zorawska E, Bodzenta-Lukaszyk A, Malyszko J. GDF-15, iron, and inflammation in early chronic kidney disease among elderly patients. Int Urol Nephrol. 2016;48(6):839–844. doi:10.1007/s11255-016-1278-z

110. Nalado AM, Olorunfemi G, Dix-Peek T, et al. Hepcidin and GDF-15 are potential biomarkers of iron deficiency anaemia in chronic kidney disease patients in South Africa. BMC Nephrol. 2020;21(1):415. doi:10.1186/s12882-020-02046-7

111. Laucyte-Cibulskiene A, Ward LJ, Ebert T, et al. Role of GDF-15, YKL-40 and MMP 9 in patients with end-stage kidney disease: focus on sex-specific associations with vascular outcomes and all-cause mortality. Biol Sex Differ. 2021;12(1):50. doi:10.1186/s13293-021-00393-0

112. Lewis GA, Rosala-Hallas A, Dodd S, et al. Characteristics associated with growth differentiation factor 15 in heart failure with preserved ejection fraction and the impact of pirfenidone. J Am Heart Assoc. 2022;11(14):e024668. doi:10.1161/JAHA.121.024668

113. Bansal N, Zelnick L, Go A, et al. Cardiac biomarkers and risk of incident heart failure in chronic kidney disease: the CRIC (Chronic Renal Insufficiency Cohort) study. J Am Heart Assoc. 2019;8(21):e012336. doi:10.1161/JAHA.119.012336

114. Natale P, Strippoli G. Sun-243 antiplatelet agents for chronic kidney disease: an updated Cochrane review. Kidney Int Rep. 2020;5(3):S299–S300. doi:10.1016/j.ekir.2020.02.778

115. Yilmaz H, Cakmak M, Darcin T, et al. Can serum Gdf-15 be associated with functional iron deficiency in hemodialysis Patients? Indian J Hematol Blood Transfus. 2016;32(2):221–227. doi:10.1007/s12288-015-0551-0

116. Shao Y, Wang H, Liu C, et al. Transforming growth factor 15 increased in severe aplastic anemia patients. Hematology. 2017;22(9):548–553. doi:10.1080/10245332.2017.1311462

117. Schechter A, Gafter-Gvili A, Shepshelovich D, et al. Post renal transplant anemia: severity, causes and their association with graft and patient survival. BMC Nephrol. 2019;20(1):51. doi:10.1186/s12882-019-1244-y

118. Thorsteinsdottir H, Salvador CL, Mjøen G, et al. Growth differentiation factor 15 in children with chronic kidney disease and after renal transplantation. Dis. Markers. 2020;2020:6162892. doi:10.1155/2020/6162892

119. de Cos Gomez M, Benito Hernandez A, Garcia Unzueta MT, et al. Growth differentiation factor 15: a biomarker with high clinical potential in the evaluation of kidney transplant candidates. J Clin Med. 2020;9(12):4112. doi:10.3390/jcm9124112

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.