Next Article in Journal
Metformin and the Liver: Unlocking the Full Therapeutic Potential
Previous Article in Journal
Selected Ion Monitoring for Orbitrap-Based Metabolomics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Brief Report

Exploring the Role of GDF-15 in Inflammatory Bowel Disease: A Case-Controlled Study Comparing Crohn’s Disease and Ulcerative Colitis with Non-Inflammatory Controls

1
Department of Medicine, First Faculty of Medicine, Charles University and Military University Hospital Prague, 169 02 Prague, Czech Republic
2
Department of Military Internal Medicine and Military Hygiene, Military Faculty of Medicine, University of Defence, 500 02 Hradec Kralove, Czech Republic
3
Department of Clinical Biochemistry, Military University Hospital, 169 02 Prague, Czech Republic
4
Department of Food Chemistry and Analysis, University of Chemistry and Technology, 160 00 Prague, Czech Republic
*
Author to whom correspondence should be addressed.
Metabolites 2024, 14(4), 185; https://doi.org/10.3390/metabo14040185
Submission received: 12 February 2024 / Revised: 8 March 2024 / Accepted: 21 March 2024 / Published: 25 March 2024

Abstract

:
Inflammatory bowel disease, encompassing Crohn’s disease and ulcerative colitis, is a persistent immune-mediated inflammatory gastrointestinal disease. This study investigates the role of growth differentiation factor 15 in severe IBD cases, aiming to identify a reliable parameter to assess disease severity and monitor activity. We analyzed plasma samples from 100 patients undergoing biologic therapy for severe IBD and 50 control subjects. Our analysis included evaluations of GDF-15 levels, inflammatory markers, and clinical features. We employed statistical methods such as the Mann–Whitney U test, ANOVA, and Spearman’s correlation for an in-depth analysis. Our results demonstrated consistently higher GDF-15 levels in patients with both Crohn’s disease and ulcerative colitis compared to the control group, irrespective of the biologic treatment received. The correlation analysis indicated significant relationships between GDF-15 levels, patient age, fibrinogen, and IL-6 levels. This study positions GDF-15 as a promising biomarker for severe IBD, with notable correlations with age and inflammatory markers. These findings underscore GDF-15’s potential in enhancing disease monitoring and management strategies in an IBD context and encourage further research to clarify GDF-15’s role in the IBD pathophysiology.

1. Introduction

Inflammatory bowel disease (IBD) is a chronic condition characterized by inflammation of the gastrointestinal tract, often accompanied by systemic inflammation and extraintestinal manifestations. It encompasses Crohn’s disease (CD) and ulcerative colitis (UC), both of which are chronic, incurable diseases with a pathogenesis that remains not fully understood. The development of these diseases involves an intricate interplay of genetic and environmental factors, which disrupt the immune system’s function. IBD commonly affects adolescents and adults, with an equal incidence in males and females. Both diseases share similar symptomatology, including abdominal pain, diarrhea, weight loss, and rectal bleeding.
Growth differentiation factor 15 (GDF-15), identified in 1997 by Bootcov et al. [1], is known by various names, including placental transforming growth factor-β (TGF-β) [2], placental bone morphogenetic protein [3], macrophage inhibitory cytokine-1 [1], and non-steroidal anti-inflammatory drug-activated gene-1 [4]. It is a peptide hormone belonging to the TGF-β superfamily [1,2] that plays critical roles in embryonic development and cellular processes such as homeostasis, cell growth, differentiation, proliferation, migration, adhesion, and apoptosis [5,6,7,8,9]. TGF-β superfamily proteins are expressed as precursors, stored in the extracellular matrix, and activated in response to stimuli like inflammation [10,11].
The GDF-15 gene is located on chromosome 19p12.1–13.1 and circulates as a 25 kDa homodimer composed of two 112 amino acid chains [12]. Under normal conditions, GDF-15 is minimally expressed in various tissues, including the lung, mammary gland, liver, pancreas, kidney, peripheral and central nervous systems, and gastrointestinal tract [13,14,15,16]. Elevated levels are observed in the placenta and prostate [17,18]. Functionally, GDF-15 acts as a hormone and a stress-induced cytokine or stress-sensitive factor [19,20], with a typical serum concentration of around 450 pg/mL [21].
Elevated GDF-15 levels have been observed in other inflammatory conditions, such as rheumatoid arthritis [22,23,24], psoriasis [25], and Behçet’s disease [26], with some studies demonstrating a correlation between serum GDF-15 levels and disease activity [23,26,27]. This study specifically aims to evaluate serum GDF-15 levels in patients with severe forms of CD and UC.

2. Materials and Methods

2.1. Study Design

In this study, we conducted a comparative analysis of plasma samples from two distinct groups: an IBD group and a control group. The IBD group comprised 100 patients diagnosed with severe CD or UC, all of whom were undergoing biologic therapy. The control group consisted of 50 individuals without any chronic inflammatory disease. To ensure the integrity of the control group, we established specific exclusion criteria, including the presence of any acute illness, pregnancy, and a history of chronic inflammatory disease, malignancy, or prostate disease. Both groups were recruited from the Military University Hospital in Prague, Czech Republic.
The primary aim of the analysis was to assess and compare the plasma levels of GDF-15 in both groups. Additionally, we sought to investigate the correlation between GDF-15 levels and various inflammatory markers, including leukocytes, high-sensitivity C-reactive protein (hsCRP), interleukin-6 (IL-6), and fibrinogen.

2.2. Sample Collection and Preparation

Blood samples for the study were collected through venipuncture during scheduled follow-up visits at the IBD Biological Treatment Center, Military University Hospital Prague, Czech Republic, between March 2022 and June 2023. Patients from both the IBD and control groups had their blood samples drawn into vacuum tubes containing EDTA. These samples were then immediately centrifuged at 3300× g for 15 min at 12 °C to obtain plasma, which was aliquoted into eight tubes per subject and stored in the dark at −80 °C until the time of analysis.
On the day of analysis, a single aliquot from each subject was thawed and brought to room temperature. After thorough mixing, the prepared samples were pipetted into labeled sample cups for analysis using a COBAS PRO analyzer (Roche, Basel, Switzerland). The results were obtained from the calibration curve, which included two-point calibration performed for the instrument, alongside the master curve provided by the device manufacturer. The total determination time for each sample was approximately 18 min, with the lowest detectable level of the assay being 400 ng/L.

2.3. Statistical Analysis

The Mann–Whitney U-test, Kruskal–Wallis test, χ2 test, and correlation and associated analyses, such as the Shapiro–Wilk test, were performed in JASP 0.18.1.0 (University of Amsterdam). The receiver operating characteristic area under the curve (ROC-AUC) was calculated in R, using the package ROCit in RStudio 2023.12.1 Build 402 with R version 4.0.2.

3. Results

3.1. Demographic and Clinical Characteristics

In this study, we enrolled 100 patients with IBD and compared their clinical parameters with those of 50 control subjects. The IBD group comprised 77 patients with severe CD and 23 patients with severe UC. All patients in the IBD group were undergoing biologic therapy, as depicted in Figure 1. Among the IBD cohort, there were 60 males (60%) and 40 females (40%), with a mean age of 43 years (standard deviation ± 9). Within this group, 18 subjects (18%) were identified as smokers. The control group included 21 males (42%) and 29 females (58%), with a similar mean age of 43 years (standard deviation ± 10); this group had four smokers (8%). A chi-squared analysis indicated a significant difference in gender distribution between the groups (p = 0.037), while differences in smoking status were not significant (χ2 = 2.66, p = 0.103). Furthermore, the Mann–Whitney U test revealed no significant age difference between the two groups (W = 2554.5, p = 0.829), as shown in Table 1, Table 2 and Table 3.

3.2. GDF-15

Our study found that the plasma GDF-15 levels were significantly higher in patients with IBD compared to the control group, as determined by the Mann–Whitney U test (p < 0.001). Further analysis using ANOVA to separately compare the Crohn’s disease group, the ulcerative colitis group, and the control group revealed consistently elevated plasma GDF-15 levels in both CD and UC (Kruskal–Wallis test, p < 0.001), as illustrated in Figure 2. Notably, the type of biologic treatment did not influence the GDF-15 levels.
In our correlation analysis using Spearman’s correlation test, we observed that increased GDF-15 levels were associated with age (ρ = 0.398, p < 0.001), fibrinogen (ρ = 0.363, p < 0.001), and IL-6 levels (ρ = 0.299, p < 0.001). However, the association of GDF-15 with hsCRP was relatively weak (ρ = 0.256, p = 0.002). An evaluation of GDF-15 as a marker to discriminate IBD patients from controls was also performed using ROC analysis, with a calculated AUC of 0.7473, indicating acceptable performance.

3.3. Conventional Markers of Inflammation

Of the serum biomarkers, the determination of CRP is the gold standard in the management of IBD patients with both symptomatic and asymptomatic disease [22]. We therefore correlated GDF-15 with CRP and other inflammatory parameters routinely measured in our hospital, such as leukocytes, fibrinogen, and IL-6. These conventional markers of inflammation were determined simultaneously with GDF-15 in both groups of patients. In the IBD group, all inflammatory markers were significantly higher than in the control group, as indicated in Table 4 and Table 5. This was despite the fact that all IBD patients were in remission at the time of blood sampling. Clinical remission is defined as the resolution of abdominal pain and the resolution of altered bowel habits [23].

4. Discussion

GDF-15, a multifunctional protein with diverse roles in health and disease, is expressed in numerous human tissues and is responsive to cellular stress. It has emerged as a significant player in various diseases, owing to its ready detectability in human plasma, which has led to its extensive utilization as a biomarker in countless studies. The associations of GDF-15 extend across a spectrum of conditions, including diabetes mellitus [24,25], cancer [26], cachexia [27], chronic inflammatory diseases [28,29,30], cognitive impairment [31,32], heart failure [33], coronary artery disease [34,35,36,37], and atrial fibrillation [38]. Its dysregulation is implicated in various pathological conditions, making it a potential therapeutic target and diagnostic biomarker for several diseases.
The exact pathophysiological effects of GDF-15 are still under intense investigation. Regarding cardiovascular effects, GDF-15 is thought to be involved in the inhibition of myocardial hypertrophy and may exert both atherogenic and antiatherogenic effects [39,40,41,42]. In the kidney, it exhibits a protective and antifibrotic effect [43,44], while, in the brain stem, it induces appetite suppression through the activation of the GFRAL receptor in the area postrema [45,46,47]. However, in the context of tumors, the data on the effect of GDF-15 are controversial, with some authors suggesting its carcinogenic activity and others pointing to its tumor suppressor activity [14,48,49,50]. Wallin et al. analyzed the expression of GDF-15 in the tumor tissue of patients with colorectal cancer. They showed that a high level of GDF-15 expression in the tumor tissue and high levels of GDF-15 in the plasma correlated with an increased risk of recurrence and reduced overall survival [51]. The study conducted by Vocka et al. [52] revealed the promising potential of GDF-15 as a biomarker in patients with metastatic colorectal cancer. Their findings demonstrated that GDF-15 exhibited comparable sensitivity to carcinoembryonic antigen (CEA) while also displaying a noteworthy correlation with liver involvement, in contrast to CEA. To the best of our knowledge, there have been no studies investigating the marker GDF-15 in non-neoplastic intestinal disorders.
Our finding aligns with several studies in rheumatoid arthritis (RA) patients, psoriasis patients, or Behçet’s disease (BD) patients, where elevated GDF-15 levels were reported [28,29,30,53,54,55,56]. Esalatmanesh et al. [53] suggested that GDF-15 levels might correlate with RA activity. Similarly, He et al. [28] observed that RA patients with positive CRP had higher plasma GDF-15 expression than those with negative CRP. In our IBD cohort, we noted a weak correlation between GDF-15 levels and both CRP and hsCRP (the hsCRP test is a highly sensitive quantification of CRP). Since CRP is a widely recognized marker of inflammation, its elevation in CD and UC typically indicates active inflammation. However, the variability in correlation strength among individuals suggests that GDF-15 might be a more reliable indicator of disease severity, activity, or the body’s response to disease.
Tasolar et al. [55] investigated GDF-15 in patients with psoriasis. The study included 50 psoriasis patients and 32 controls. The psoriasis patients were divided into three groups based on the Psoriasis Area Severity Index score (PASI, a tool used to measure the severity and extent of psoriasis): patients with mild psoriasis (PASI < 10), patients with moderate psoriasis (PASI 10–20), and patients with severe psoriasis (PASI > 20). No or statistically insignificant differences were observed between psoriasis patients and healthy controls for interleukin-12 (IL-12), interleukin-17a (IL-17a), interleukin-22 (IL-22), and interleukin-23 (IL-23). In contrast, a statistically significant difference was observed for hsCRP, tumor necrosis factor-alpha (TNF-α), and GDF-15. GDF-15 was significantly higher in all three patient groups (mild, moderate, severe psoriasis). Furthermore, this study confirmed the strong correlation between the disease duration and PASI score. A serum GDF-15 level above 1498 pg/mL was identified as potentially predictive of a high PASI score. Akbari et al.’s case–control study compared 45 psoriatic patients with 45 healthy individuals. They demonstrated a significant association between serum GDF-15 levels and psoriasis, as well as between GDF-15 gene expression and psoriasis. Both serum GDF-15 levels and GDF-15 gene expression correlated with disease severity [29].
Elbarky et al. [30] compared serum GDF-15 levels in 30 patients with BD and 20 controls. Sarıyıldız et al. [56] compared serum GDF-15 levels in 46 patients with BD and 30 healthy subjects. In both studies, the BD patients were divided into two groups according to the presence of peripheral arthritis and arthralgia. In both studies, the authors reached similar conclusions, showing that BD patients with peripheral arthritis and arthralgia had significantly higher serum GDF-15 levels than patients without peripheral arthritis or controls. These findings may be explained by the increased GDF-15 expression in activated macrophages after stimulation with proinflammatory cytokines such as TNF-α, interleukin-1β (IL-1β), and IL-6 in the synovium during inflammation [40]. Therefore, GDF-15 could be used as a marker for peripheral arthritis in BD patients.
A team of authors from the University of Colorado demonstrated significantly elevated plasma levels of GDF-15 in patients with systemic sclerosis-associated pulmonary arterial hypertension (SSc-PAH) compared to SSc patients without PAH. The GDF-15 levels were positively correlated with echocardiographic estimates of right ventricular systolic pressure and plasma levels of the amino-terminal propeptide form of brain natriuretic peptide (NT-proBNP). Plasma GDF-15 levels greater than 125 pg/mL were associated with reduced survival. Additionally, the expression of the GDF-15 protein was found to be increased in the lung tissue of patients with SSc-PAH [57].
CRP is the most commonly used serum biomarker of inflammation in IBD patients. However, CRP is not disease-specific [58]. In patients with CD who are in symptomatic remission, CRP levels below 5 mg/L indicate the absence of active inflammation [22]. In their investigation, Con et al. [59] examined the dynamics of C-reactive protein (CRP) in individuals with acute severe ulcerative colitis (UC) following the administration of infliximab as a salvage therapy. This comprehensive study included a cohort of 94 patients, 20% of whom required colectomy within 12 months. At the end of the study, patients were stratified into two distinct categories—those who did not require colectomy and those who did. Notably, the baseline CRP levels showed no statistically significant difference between the two cohorts (colectomy group: median 65 mg/L, non-colectomy group: median 46 mg/L). On the first day after infliximab administration, CRP levels showed a significant decrease in both groups (colectomy group: median 32 mg/L, non-colectomy group: median 11 mg/L). On the third day after infliximab administration, the median CRP levels continued to decrease to 17 mg/L and 5 mg/L, respectively.
Considering these findings, the monitoring of GDF-15 levels alongside traditional markers like CRP, complete blood counts, or fecal calprotectin could offer a more comprehensive assessment of the inflammatory status in IBD patients, thereby enhancing the disease management strategies. Future longitudinal studies should focus on varying treatment modalities, such as aminosalicylates, corticosteroids, and immunosuppressants, to understand how GDF-15 levels evolve with different therapeutic interventions. This approach could reveal patterns linked to the treatment response and disease progression. Comparisons of the GDF-15 levels between biologic-naive patients and those on various biologic regimens could elucidate the biologics’ specific impact on GDF-15 and its potential as a marker for the treatment response.
By monitoring the GDF-15 levels at strategic time points, we could assess its value as a predictive marker for disease flare-ups or complications in IBD patients. This would have significant implications in terms of tailoring individual treatment plans and facilitating early intervention, ultimately improving patient outcomes.
Overall, while CRP is a valuable biomarker in IBD management, GDF-15 may potentially offer certain advantages in terms of specificity, early detection, correlations with disease activity, and predictive value. However, further research is needed to fully validate its utility and establish its clinical significance in routine practice.

5. Study Limitations

  • This was a small, single-center, cross-sectional study designed to compare the plasma GDF-15 levels between patients with CD and UC receiving biologic therapy and a control group.
  • The study primarily focused on patients with severe IBD undergoing biologic therapy, which may limit the generalizability of the findings to patients with less severe forms of IBD or those not receiving biologic treatment.
  • While the study identified associations between plasma GDF-15 levels and severe IBD, the clinical significance of these findings in terms of diagnosis, prognosis, or treatment remains to be fully elucidated.

6. Conclusions

  • Patients with severe IBD, including CD and UC, undergoing biologic therapy, exhibited significantly higher levels of GDF-15 in their plasma compared to individuals without chronic inflammatory diseases.
  • There was a positive correlation between elevated GDF-15 levels and other markers of inflammation, such as age, fibrinogen, and IL-6, indicating that GDF-15 may serve as a complementary marker in assessing the disease status in IBD patients.
  • Further research is warranted to uncover the specific mechanisms underlying GDF-15 dysregulation in IBD and to explore its potential as a biomarker for disease activity and the treatment response.

Author Contributions

Conceptualization, O.K. and M.M.; methodology, M.B. and V.K.; software, V.K.; validation, P.M. and J.M.H.; formal analysis, V.K.; investigation, O.K. and P.M.; resources, O.K. and P.M.; writing—original draft preparation, O.K.; writing—review and editing, J.M.H. and M.M.; supervision, P.U. and M.M.; funding acquisition, J.M.H. and P.U. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Education, Youth and Sports of the Czech Republic—Specific research SV/FVZ202201 (Military Faculty of Medicine, University of Defence, Hradec Kralove, Czech Republic) and by the Ministry of Defence of the Czech Republic, projects MO 1012 (Military University Hospital Prague, Czech Republic) and MO 1011—Clinical Fields II (Military Faculty of Medicine, University of Defence, Hradec Kralove, Czech Republic).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of the Military University Hospital in Prague, Czech Republic (protocol code 108/17-25/2022, date of approval 21 March 2022).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available because they contain information that could compromise the privacy of the research participants.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Bootcov, M.R.; Bauskin, A.R.; Valenzuela, S.M.; Moore, A.G.; Bansal, M.; He, X.Y.; Zhang, H.P.; Donnellan, M.; Mahler, S.; Pryor, K.; et al. MIC-1, a Novel Macrophage Inhibitory Cytokine, Is a Divergent Member of the TGF-β Superfamily. Proc. Natl. Acad. Sci. USA 1997, 94, 11514–11519. [Google Scholar] [CrossRef]
  2. Lawton, L.N.; Bonaldo, M.D.F.; Jelenc, P.C.; Qiu, L.; Baumes, S.A.; Marcelino, R.A.; De Jesus, G.M.; Wellington, S.; Knowles, J.A.; Warburton, D.; et al. Identification of a Novel Member of the TGF-Beta Superfamily Highly Expressed in Human Placenta. Gene 1997, 203, 17–26. [Google Scholar] [CrossRef] [PubMed]
  3. Hromas, R.; Hufford, M.; Sutton, J.; Xu, D.; Li, Y.; Lu, L. PLAB, a Novel Placental Bone Morphogenetic Protein. Biochim. Biophys. Acta (BBA) Gene Struct. Expr. 1997, 1354, 40–44. [Google Scholar] [CrossRef]
  4. Baek, S.J.; Horowitz, J.M.; Eling, T.E. Molecular Cloning and Characterization of Human Nonsteroidal Anti-Inflammatory Drug-Activated Gene Promoter. J. Biol. Chem. 2001, 276, 33384–33392. [Google Scholar] [CrossRef] [PubMed]
  5. Gordon, K.J.; Blobe, G.C. Role of Transforming Growth Factor-β Superfamily Signaling Pathways in Human Disease. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2008, 1782, 197–228. [Google Scholar] [CrossRef] [PubMed]
  6. Santibañez, J.F.; Quintanilla, M.; Bernabeu, C. TGF-β/TGF-β Receptor System and Its Role in Physiological and Pathological Conditions. Clin. Sci. 2011, 121, 233–251. [Google Scholar] [CrossRef] [PubMed]
  7. De Caestecker, M. The Transforming Growth Factor-β Superfamily of Receptors. Cytokine Growth Factor Rev. 2004, 15, 1–11. [Google Scholar] [CrossRef] [PubMed]
  8. Siegel, P.M.; Massagué, J. Cytostatic and Apoptotic Actions of TGF-β in Homeostasis and Cancer. Nat. Rev. Cancer 2003, 3, 807–820. [Google Scholar] [CrossRef]
  9. Battegay, E.J.; Raines, E.W.; Seifert, R.A.; Bowen-Pope, D.F.; Ross, R. TGF-β Induces Bimodal Proliferation of Connective Tissue Cells via Complex Control of an Autocrine PDGF Loop. Cell 1990, 63, 515–524. [Google Scholar] [CrossRef]
  10. Xu, X.; Zheng, L.; Yuan, Q.; Zhen, G.; Crane, J.L.; Zhou, X.; Cao, X. Transforming Growth Factor-β in Stem Cells and Tissue Homeostasis. Bone Res. 2018, 6, 2. [Google Scholar] [CrossRef]
  11. Assadi, A.; Zahabi, A.; Hart, R.A. GDF15, an Update of the Physiological and Pathological Roles It Plays: A Review. Pflug. Arch. Eur. J. Physiol. 2020, 472, 1535–1546. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, D.; Day, E.A.; Townsend, L.K.; Djordjevic, D.; Jørgensen, S.B.; Steinberg, G.R. GDF15: Emerging Biology and Therapeutic Applications for Obesity and Cardiometabolic Disease. Nat. Rev. Endocrinol. 2021, 17, 592–607. [Google Scholar] [CrossRef] [PubMed]
  13. Böttner, M.; Laaff, M.; Schechinger, B.; Rappold, G.; Unsicker, K.; Suter-Crazzolara, C. Characterization of the Rat, Mouse, and Human Genes of Growth/Differentiation Factor-15/Macrophage Inhibiting Cytokine-1 (GDF-15/MIC-1). Gene 1999, 237, 105–111. [Google Scholar] [CrossRef] [PubMed]
  14. Mimeault, M.; Batra, S.K. Divergent Molecular Mechanisms Underlying the Pleiotropic Functions of Macrophage Inhibitory Cytokine-1 in Cancer. J. Cell. Physiol. 2010, 224, 626–635. [Google Scholar] [CrossRef] [PubMed]
  15. Koopmann, J.; Buckhaults, P.; Brown, D.A.; Zahurak, M.L.; Sato, N.; Fukushima, N.; Sokoll, L.J.; Chan, D.W.; Yeo, C.J.; Hruban, R.H.; et al. Serum Macrophage Inhibitory Cytokine 1 as a Marker of Pancreatic and Other Periampullary Cancers. Clin. Cancer Res. 2004, 10, 2386–2392. [Google Scholar] [CrossRef] [PubMed]
  16. Strelau, J.; Strzelczyk, A.; Rusu, P.; Bendner, G.; Wiese, S.; Diella, F.; Altick, A.L.; Von Bartheld, C.S.; Klein, R.; Sendtner, M.; et al. Progressive Postnatal Motoneuron Loss in Mice Lacking GDF-15. J. Neurosci. 2009, 29, 13640–13648. [Google Scholar] [CrossRef] [PubMed]
  17. Lambert, J.R.; Kelly, J.A.; Shim, M.; Huffer, W.E.; Nordeen, S.K.; Baek, S.J.; Eling, T.E.; Lucia, M.S. Prostate Derived Factor in Human Prostate Cancer Cells: Gene Induction by Vitamin D via a P53-dependent Mechanism and Inhibition of Prostate Cancer Cell Growth. J. Cell. Physiol. 2006, 208, 566–574. [Google Scholar] [CrossRef]
  18. Yokoyama-Kobayashi, M.; Saeki, M.; Sekine, S.; Kato, S. Human cDNA Encoding a Novel TGF- Superfamily Protein Highly Expressed in Placenta. J. Biochem. 1997, 122, 622–626. [Google Scholar] [CrossRef]
  19. Kim, K.H.; Kim, S.H.; Han, D.H.; Jo, Y.S.; Lee, Y.; Lee, M.-S. Growth Differentiation Factor 15 Ameliorates Nonalcoholic Steatohepatitis and Related Metabolic Disorders in Mice. Sci. Rep. 2018, 8, 6789. [Google Scholar] [CrossRef]
  20. Kleinert, M.; Clemmensen, C.; Sjøberg, K.A.; Carl, C.S.; Jeppesen, J.F.; Wojtaszewski, J.F.P.; Kiens, B.; Richter, E.A. Exercise Increases Circulating GDF15 in Humans. Mol. Metab. 2018, 9, 187–191. [Google Scholar] [CrossRef]
  21. Tsai, V.W.-W.; Macia, L.; Feinle-Bisset, C.; Manandhar, R.; Astrup, A.; Raben, A.; Lorenzen, J.K.; Schmidt, P.T.; Wiklund, F.; Pedersen, N.L.; et al. Serum Levels of Human MIC-1/GDF15 Vary in a Diurnal Pattern, Do Not Display a Profile Suggestive of a Satiety Factor and Are Related to BMI. PLoS ONE 2015, 10, e0133362. [Google Scholar] [CrossRef] [PubMed]
  22. Ananthakrishnan, A.N.; Adler, J.; Chachu, K.A.; Nguyen, N.H.; Siddique, S.M.; Weiss, J.M.; Sultan, S.; Velayos, F.S.; Cohen, B.L.; Singh, S. AGA Clinical Practice Guideline on the Role of Biomarkers for the Management of Crohn’s Disease. Gastroenterology 2023, 165, 1367–1399. [Google Scholar] [CrossRef] [PubMed]
  23. Peyrin-Biroulet, L.; Sandborn, W.; Sands, B.E.; Reinisch, W.; Bemelman, W.; Bryant, R.V.; D’Haens, G.; Dotan, I.; Dubinsky, M.; Feagan, B.; et al. Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE): Determining Therapeutic Goals for Treat-to-Target. Am. J. Gastroenterol. 2015, 110, 1324–1338. [Google Scholar] [CrossRef] [PubMed]
  24. Gerstein, H.C.; Pare, G.; Hess, S.; Ford, R.J.; Sjaarda, J.; Raman, K.; McQueen, M.; Lee, S.; Haenel, H.; Steinberg, G.R. Growth Differentiation Factor 15 as a Novel Biomarker for Metformin. Diabetes Care 2017, 40, 280–283. [Google Scholar] [CrossRef]
  25. Al-kuraishy, H.M.; Al-Gareeb, A.I.; Alexiou, A.; Papadakis, M.; Nadwa, E.H.; Albogami, S.M.; Alorabi, M.; Saad, H.M.; Batiha, G.E. Metformin and Growth Differentiation Factor 15 (GDF15) in Type 2 Diabetes Mellitus: A Hidden Treasure. J. Diabetes 2022, 14, 806–814. [Google Scholar] [CrossRef]
  26. Welsh, J.B.; Sapinoso, L.M.; Kern, S.G.; Brown, D.A.; Liu, T.; Bauskin, A.R.; Ward, R.L.; Hawkins, N.J.; Quinn, D.I.; Russell, P.J.; et al. Large-Scale Delineation of Secreted Protein Biomarkers Overexpressed in Cancer Tissue and Serum. Proc. Natl. Acad. Sci. USA 2003, 100, 3410–3415. [Google Scholar] [CrossRef] [PubMed]
  27. Johnen, H.; Lin, S.; Kuffner, T.; Brown, D.A.; Tsai, V.W.-W.; Bauskin, A.R.; Wu, L.; Pankhurst, G.; Jiang, L.; Junankar, S.; et al. Tumor-Induced Anorexia and Weight Loss Are Mediated by the TGF-β Superfamily Cytokine MIC-1. Nat. Med. 2007, 13, 1333–1340. [Google Scholar] [CrossRef]
  28. He, Y.-W.; He, C.-S. Association of Growth and Differentiation Factor 15 in Rheumatoid Arthritis. J. Inflamm. Res. 2022, 15, 1173–1181. [Google Scholar] [CrossRef] [PubMed]
  29. Akbari, H.; Talaee, R.; Zaker, S.F.; Nikoueinejad, H. Investigating the Correlation between Growth Differentiation Factor 15 Serum Level and Its Gene Expression with Psoriasis and Its Severity. Iran. J. Allergy Asthma Immunol. 2021, 20, 593–599. [Google Scholar] [CrossRef]
  30. Elbarky, E.M.; Hussien, M.I.; Elgazzar, N.M.; Mabrouk, M.M.; Elsaadany, H.M. Serum Growth Differentiation Factor-15 (GDF-15) Level in Behcet’s Disease Patients: Relation to Clinical Characteristics, Musculoskeletal Ultrasound Findings and Disease Activity. Egypt. Rheumatol. 2021, 43, 261–266. [Google Scholar] [CrossRef]
  31. Fuchs, T.; Trollor, J.N.; Crawford, J.; Brown, D.A.; Baune, B.T.; Samaras, K.; Campbell, L.; Breit, S.N.; Brodaty, H.; Sachdev, P.; et al. Macrophage Inhibitory Cytokine-1 Is Associated with Cognitive Impairment and Predicts Cognitive Decline—The Sydney Memory and Aging Study. Aging Cell 2013, 12, 882–889. [Google Scholar] [CrossRef] [PubMed]
  32. Chai, Y.L.; Hilal, S.; Chong, J.P.C.; Ng, Y.X.; Liew, O.W.; Xu, X.; Ikram, M.K.; Venketasubramanian, N.; Richards, A.M.; Lai, M.K.P.; et al. Growth Differentiation Factor-15 and White Matter Hyperintensities in Cognitive Impairment and Dementia. Medicine 2016, 95, e4566. [Google Scholar] [CrossRef] [PubMed]
  33. Bouabdallaoui, N.; Claggett, B.; Zile, M.R.; McMurray, J.J.V.; O’Meara, E.; Packer, M.; Prescott, M.F.; Swedberg, K.; Solomon, S.D.; Rouleau, J.L.; et al. Growth Differentiation Factor-15 Is Not Modified by Sacubitril/Valsartan and Is an Independent Marker of Risk in Patients with Heart Failure and Reduced Ejection Fraction: The PARADIGM-HF Trial. Eur. J. Heart Fail. 2018, 20, 1701–1709. [Google Scholar] [CrossRef] [PubMed]
  34. Kempf, T.; Bjorklund, E.; Olofsson, S.; Lindahl, B.; Allhoff, T.; Peter, T.; Tongers, J.; Wollert, K.C.; Wallentin, L. Growth-Differentiation Factor-15 Improves Risk Stratification in ST-Segment Elevation Myocardial Infarction. Eur. Heart J. 2007, 28, 2858–2865. [Google Scholar] [CrossRef] [PubMed]
  35. Wollert, K.C.; Kempf, T.; Peter, T.; Olofsson, S.; James, S.; Johnston, N.; Lindahl, B.; Horn-Wichmann, R.; Brabant, G.; Simoons, M.L.; et al. Prognostic Value of Growth-Differentiation Factor-15 in Patients With Non–ST-Elevation Acute Coronary Syndrome. Circulation 2007, 115, 962–971. [Google Scholar] [CrossRef] [PubMed]
  36. Lindholm, D.; James, S.K.; Gabrysch, K.; Storey, R.F.; Himmelmann, A.; Cannon, C.P.; Mahaffey, K.W.; Steg, P.G.; Held, C.; Siegbahn, A.; et al. Association of Multiple Biomarkers With Risk of All-Cause and Cause-Specific Mortality After Acute Coronary Syndromes: A Secondary Analysis of the PLATO Biomarker Study. JAMA Cardiol. 2018, 3, 1160. [Google Scholar] [CrossRef]
  37. Li, M.; Duan, L.; Cai, Y.-L.; Li, H.-Y.; Hao, B.-C.; Chen, J.-Q.; Liu, H.-B. Growth Differentiation Factor-15 Is Associated with Cardiovascular Outcomes in Patients with Coronary Artery Disease. Cardiovasc. Diabetol. 2020, 19, 120. [Google Scholar] [CrossRef]
  38. Sharma, A.; Hijazi, Z.; Andersson, U.; Al-Khatib, S.M.; Lopes, R.D.; Alexander, J.H.; Held, C.; Hylek, E.M.; Leonardi, S.; Hanna, M.; et al. Use of Biomarkers to Predict Specific Causes of Death in Patients With Atrial Fibrillation: Insights From the ARISTOTLE Trial. Circulation 2018, 138, 1666–1676. [Google Scholar] [CrossRef]
  39. Xu, J.; Kimball, T.R.; Lorenz, J.N.; Brown, D.A.; Bauskin, A.R.; Klevitsky, R.; Hewett, T.E.; Breit, S.N.; Molkentin, J.D. GDF15/MIC-1 Functions As a Protective and Antihypertrophic Factor Released From the Myocardium in Association With SMAD Protein Activation. Circ. Res. 2006, 98, 342–350. [Google Scholar] [CrossRef]
  40. Bonaterra, G.A.; Zügel, S.; Thogersen, J.; Walter, S.A.; Haberkorn, U.; Strelau, J.; Kinscherf, R. Growth Differentiation Factor-15 Deficiency Inhibits Atherosclerosis Progression by Regulating Interleukin-6–Dependent Inflammatory Response to Vascular Injury. J. Am. Heart Assoc. 2012, 1, e002550. [Google Scholar] [CrossRef]
  41. Preusch, M.R.; Baeuerle, M.; Albrecht, C.; Blessing, E.; Bischof, M.; Katus, H.A.; Bea, F. GDF-15 Protects from Macrophage Accumulation in a Mousemodel of Advanced Atherosclerosis. Eur. J. Med. Res. 2013, 18, 19. [Google Scholar] [CrossRef] [PubMed]
  42. Johnen, H.; Kuffner, T.; Brown, D.A.; Wu, B.J.; Stocker, R.; Breit, S.N. Increased Expression of the TGF-b Superfamily Cytokine MIC-1/GDF15 Protects ApoE−/− Mice from the Development of Atherosclerosis. Cardiovasc. Pathol. 2012, 21, 499–505. [Google Scholar] [CrossRef] [PubMed]
  43. Nair, V.; Robinson-Cohen, C.; Smith, M.R.; Bellovich, K.A.; Bhat, Z.Y.; Bobadilla, M.; Brosius, F.; De Boer, I.H.; Essioux, L.; Formentini, I.; et al. Growth Differentiation Factor–15 and Risk of CKD Progression. J. Am. Soc. Nephrol. 2017, 28, 2233–2240. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, Y.-I.; Shin, H.-W.; Chun, Y.-S.; Park, J.-W. CST3 and GDF15 Ameliorate Renal Fibrosis by Inhibiting Fibroblast Growth and Activation. Biochem. Biophys. Res. Commun. 2018, 500, 288–295. [Google Scholar] [CrossRef] [PubMed]
  45. Emmerson, P.J.; Wang, F.; Du, Y.; Liu, Q.; Pickard, R.T.; Gonciarz, M.D.; Coskun, T.; Hamang, M.J.; Sindelar, D.K.; Ballman, K.K.; et al. The Metabolic Effects of GDF15 Are Mediated by the Orphan Receptor GFRAL. Nat. Med. 2017, 23, 1215–1219. [Google Scholar] [CrossRef] [PubMed]
  46. Mullican, S.E.; Lin-Schmidt, X.; Chin, C.-N.; Chavez, J.A.; Furman, J.L.; Armstrong, A.A.; Beck, S.C.; South, V.J.; Dinh, T.Q.; Cash-Mason, T.D.; et al. GFRAL Is the Receptor for GDF15 and the Ligand Promotes Weight Loss in Mice and Nonhuman Primates. Nat. Med. 2017, 23, 1150–1157. [Google Scholar] [CrossRef]
  47. Yang, L.; Chang, C.-C.; Sun, Z.; Madsen, D.; Zhu, H.; Padkjær, S.B.; Wu, X.; Huang, T.; Hultman, K.; Paulsen, S.J.; et al. GFRAL Is the Receptor for GDF15 and Is Required for the Anti-Obesity Effects of the Ligand. Nat. Med. 2017, 23, 1158–1166. [Google Scholar] [CrossRef] [PubMed]
  48. Bauskin, A.R.; Brown, D.A.; Kuffner, T.; Johnen, H.; Luo, X.W.; Hunter, M.; Breit, S.N. Role of Macrophage Inhibitory Cytokine-1 in Tumorigenesis and Diagnosis of Cancer. Cancer Res. 2006, 66, 4983–4986. [Google Scholar] [CrossRef]
  49. Siddiqui, J.A.; Pothuraju, R.; Khan, P.; Sharma, G.; Muniyan, S.; Seshacharyulu, P.; Jain, M.; Nasser, M.W.; Batra, S.K. Pathophysiological Role of Growth Differentiation Factor 15 (GDF15) in Obesity, Cancer, and Cachexia. Cytokine Growth Factor Rev. 2022, 64, 71–83. [Google Scholar] [CrossRef]
  50. Corre, J.; Hébraud, B.; Bourin, P. Concise Review: Growth Differentiation Factor 15 in Pathology: A Clinical Role? Stem Cells Transl. Med. 2013, 2, 946–952. [Google Scholar] [CrossRef]
  51. Wallin, U.; Glimelius, B.; Jirström, K.; Darmanis, S.; Nong, R.Y.; Pontén, F.; Johansson, C.; Påhlman, L.; Birgisson, H. Growth Differentiation Factor 15: A Prognostic Marker for Recurrence in Colorectal Cancer. Br. J. Cancer 2011, 104, 1619–1627. [Google Scholar] [CrossRef] [PubMed]
  52. Vocka, M.; Langer, D.; Fryba, V.; Petrtyl, J.; Hanus, T.; Kalousova, M.; Zima, T.; Petruzelka, L. Growth/Differentiation Factor 15 (GDF-15) as New Potential Serum Marker in Patients with Metastatic Colorectal Cancer. Cancer Biomark. 2018, 21, 869–874. [Google Scholar] [CrossRef] [PubMed]
  53. Esalatmanesh, K.; Fayyazi, H.; Esalatmanesh, R.; Khabbazi, A. The Association between Serum Levels of Growth Differentiation Factor-15 and Rheumatoid Arthritis Activity. Int. J. Clin. Pract. 2020, 74, e13564. [Google Scholar] [CrossRef] [PubMed]
  54. Khalil, M.E.; Elhanafy, M.S.; Eigela, S.E.; Nasr, H.E.; ELgendy, M.E. Serum GDF-15 Level in Rheumatoid Arthritis and Relation to Disease Activity and Severity. Benha J. Appl. Sci. 2020, 5, 131–134. [Google Scholar] [CrossRef]
  55. Taşolar, M.K.; Erfan, G.; Raimoğlu, O.; Albayrak, H.; Yanık, M.E. Role of GDF-15 as an Inflammatory Marker in Patients with Psoriasis Vulgaris. Arch. Turk. Dermatol. Venerol. 2021, 55, 184–188. [Google Scholar] [CrossRef]
  56. Sarıyıldız, M.A.; Yazmalar, L.; Batmaz, İ.; Alpaycı, M.; Burkan, Y.K.; Sula, B.; Kaplan, İ.; Yıldız, M.; Akar, Z.A.; Bozkurt, M. Serum GDF -15 Level in Behçet’s Disease: Relationships between Disease Activity and Clinical Parameters. Int. J. Dermatol. 2016, 55, 1289–1294. [Google Scholar] [CrossRef] [PubMed]
  57. Meadows, C.A.; Risbano, M.G.; Zhang, L.; Geraci, M.W.; Tuder, R.M.; Collier, D.H.; Bull, T.M. Increased Expression of Growth Differentiation Factor-15 in Systemic Sclerosis-Associated Pulmonary Arterial Hypertension. Chest 2011, 139, 994–1002. [Google Scholar] [CrossRef]
  58. Chen, P.; Zhou, G.; Lin, J.; Li, L.; Zeng, Z.; Chen, M.; Zhang, S. Serum Biomarkers for Inflammatory Bowel Disease. Front. Med. 2020, 7, 123. [Google Scholar] [CrossRef]
  59. Con, D.; Andrew, B.; Nicolaides, S.; Van Langenberg, D.R.; Vasudevan, A. Biomarker Dynamics during Infliximab Salvage for Acute Severe Ulcerative Colitis: C-Reactive Protein (CRP)-Lymphocyte Ratio and CRP-Albumin Ratio Are Useful in Predicting Colectomy. Intestig. Res. 2022, 20, 101–113. [Google Scholar] [CrossRef]
Figure 1. Number of patients by type of biological treatment.
Figure 1. Number of patients by type of biological treatment.
Metabolites 14 00185 g001
Figure 2. (a) GDF-15 levels in the IBD group and the non-inflammatory control group; (b) GDF-15 levels in patients with Crohn’s disease (Crohn), patients with ulcerative colitis (UC), and patients without inflammatory diseases (control).
Figure 2. (a) GDF-15 levels in the IBD group and the non-inflammatory control group; (b) GDF-15 levels in patients with Crohn’s disease (Crohn), patients with ulcerative colitis (UC), and patients without inflammatory diseases (control).
Metabolites 14 00185 g002
Table 1. Patient characteristics.
Table 1. Patient characteristics.
CharacteristicsIBD PatientsControlsp-ValueTest
n = 100n = 50
Age (y)43 (25–67)43 (18–61)0.829U-test
Sex (male)60 (60%)21 (42%)0.037χ2
Sex (female)40 (40%)29 (58%)0.037χ2
Current smoker18 (18%)4 (8%)0.103χ2
Body mass index25
Total protein (g/L)73.6710.003t-test
Albumin (g/L)44.244.60.449t-test
Abdominal surgery71 (71%)
Disease duration (y)13.4
Table 2. Number of patients by disease phenotype.
Table 2. Number of patients by disease phenotype.
CDn = 77
inflammatory38
stricturing10
fistulizing29
UCn = 23
extensive colitis17
left-sided colitis5
Table 3. Treatment of IBD patients and their chronic inflammatory comorbidities.
Table 3. Treatment of IBD patients and their chronic inflammatory comorbidities.
Type of TreatmentNumber of Patients
biologic therapy100 (100%)
5-aminosalicylic acid (5-ASA)63 (63%)
azathioprine8 (8%)
methotrexate1 (1%)
corticoids2 (2%)
Concomitant chronic inflammatory diseasesNumber of patients
autoimmune thyroiditis2
celiac disease1
psoriasis5
hidradenitis suppurativa2
atopic eczema2
antiphospholipid syndrome1
Table 4. GDF-15 and inflammatory markers.
Table 4. GDF-15 and inflammatory markers.
MedianMeanStd. DeviationMinimumMaximum
GDF-15 (ng/L)Control527569.34188.6554001171
IBD751943.23608.5794003406
Leukocytes (×109/L)Control6.0956.3381.7033.4411.58
IBD6.847.1091.876313.17
hsCRP (µg/L)Control817.552113.3222611.358259461
IBD1511.72977.5223811.36587.624,497
IL-6 (ng/L)Control1.51.7760.6171.54.3
IBD1.62.7512.5561.518
Fibrinogen (g/L)Control2.622.6680.4831.774.04
IBD2.822.8610.4741.84.11
Table 5. Inflammatory markers.
Table 5. Inflammatory markers.
Mann-Whitney U Test.
Wp
GDF-151263.5<0.001
Leukocytes1867.50.012
hsCRP19700.035
IL-61825.50.003
Fibrinogen1902.50.017
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kučerka, O.; Blahutová, M.; Kosek, V.; Mináriková, P.; Horáček, J.M.; Urbánek, P.; Malý, M. Exploring the Role of GDF-15 in Inflammatory Bowel Disease: A Case-Controlled Study Comparing Crohn’s Disease and Ulcerative Colitis with Non-Inflammatory Controls. Metabolites 2024, 14, 185. https://doi.org/10.3390/metabo14040185

AMA Style

Kučerka O, Blahutová M, Kosek V, Mináriková P, Horáček JM, Urbánek P, Malý M. Exploring the Role of GDF-15 in Inflammatory Bowel Disease: A Case-Controlled Study Comparing Crohn’s Disease and Ulcerative Colitis with Non-Inflammatory Controls. Metabolites. 2024; 14(4):185. https://doi.org/10.3390/metabo14040185

Chicago/Turabian Style

Kučerka, Ondřej, Marie Blahutová, Vít Kosek, Petra Mináriková, Jan M. Horáček, Petr Urbánek, and Martin Malý. 2024. "Exploring the Role of GDF-15 in Inflammatory Bowel Disease: A Case-Controlled Study Comparing Crohn’s Disease and Ulcerative Colitis with Non-Inflammatory Controls" Metabolites 14, no. 4: 185. https://doi.org/10.3390/metabo14040185

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop