Proteomic profiles before and during weight loss: Results from randomized trial of dietary intervention

Sylwia M Figarska, Joseph Rigdon, Andrea Ganna, Sölve Elmståhl, Lars Lind, Christopher D Gardner, Erik Ingelsson, Sylwia M Figarska, Joseph Rigdon, Andrea Ganna, Sölve Elmståhl, Lars Lind, Christopher D Gardner, Erik Ingelsson

Abstract

Inflammatory and cardiovascular biomarkers have been associated with obesity, but little is known about how they change upon dietary intervention and concomitant weight loss. Further, protein biomarkers might be useful for predicting weight loss in overweight and obese individuals. We performed secondary analyses in the Diet Intervention Examining The Factors Interacting with Treatment Success (DIETFITS) randomized intervention trial that included healthy 609 adults (18-50 years old) with BMI 28-40 kg/m2, to evaluate associations between circulating protein biomarkers and BMI at baseline, during a weight loss diet intervention, and to assess predictive potential of baseline blood proteins on weight loss. We analyzed 263 plasma proteins at baseline and 6 months into the intervention using the Olink Proteomics CVD II, CVD III and Inflammation arrays. BMI was assessed at baseline, after 3 and 6 months of dietary intervention. At baseline, 102 of the examined inflammatory and cardiovascular biomarkers were associated with BMI (>90% with successful replication in 1,584 overweight/obese individuals from a community-based cohort study) and 130 tracked with weight loss shedding light into the pathophysiology of obesity. However, out of 263 proteins analyzed at baseline, only fibroblast growth factor 21 (FGF-21) predicted weight loss, and none helped individualize dietary assignment.

Conflict of interest statement

Erik Ingelsson received consulting fees from Olink Proteomics (in 2017–2018) for work unrelated to the present project. The company had no influence over design, analysis or interpretation of data in the present study, and did not provide any funding for the study. The remaining authors do not have any conflict of interest.

Figures

Figure 1
Figure 1
Associations between cross-sectional associations of protein levels and BMI at baseline in DIETFITS. Proteins significantly associated with BMI (at FDR 

Figure 2

Cross-sectional associations between proteins and…

Figure 2

Cross-sectional associations between proteins and BMI at baseline (left panel); and associations between…

Figure 2
Cross-sectional associations between proteins and BMI at baseline (left panel); and associations between changes in proteins and changes in BMI during 6 months (right panel). Results are shown for 93 proteins significantly associated in both analyses. Red colors indicate positive coefficients (of baseline levels or changes, respectively), while blue colors indicate negative coefficients.
Figure 2
Figure 2
Cross-sectional associations between proteins and BMI at baseline (left panel); and associations between changes in proteins and changes in BMI during 6 months (right panel). Results are shown for 93 proteins significantly associated in both analyses. Red colors indicate positive coefficients (of baseline levels or changes, respectively), while blue colors indicate negative coefficients.

References

    1. Hagg S, et al. Adiposity as a cause of cardiovascular disease: a Mendelian randomization study. Int. J. Epidemiol. 2015;44:578–586. doi: 10.1093/ije/dyv094.
    1. Holmes MV, et al. Causal effects of body mass index on cardiometabolic traits and events: a Mendelian randomization analysis. Am. J. Hum. Genet. 2014;94:198–208. doi: 10.1016/j.ajhg.2013.12.014.
    1. Wang T, et al. Causal Association of Overall Obesity and Abdominal Obesity with Type 2 Diabetes: A Mendelian Randomization Analysis. Obesity. 2018;26:934–942. doi: 10.1002/oby.22167.
    1. Aleksandrova K, Mozaffarian D, Pischon T. Addressing the Perfect Storm: Biomarkers in Obesity and Pathophysiology of Cardiometabolic Risk. Clin. Chem. 2018;64:142–153. doi: 10.1373/clinchem.2017.275172.
    1. Piening BD, et al. Integrative Personal Omics Profiles during Periods of Weight Gain and Loss. Cell Syst. 2018;6:157–170.e158. doi: 10.1016/j.cels.2017.12.013.
    1. Soare A, Weiss EP, Pozzilli P. Benefits of caloric restriction for cardiometabolic health, including type 2 diabetes mellitus risk. Diabetes Metab. Res. Rev. 2014;30(Suppl 1):41–47. doi: 10.1002/dmrr.2517.
    1. Alves NE, Enes BN, Martino HS, Alfenas Rde C, Ribeiro SM. Meal replacement based on Human Ration modulates metabolic risk factors during body weight loss: a randomized controlled trial. Eur. J. Nutr. 2014;53:939–950. doi: 10.1007/s00394-013-0598-3.
    1. Armenise C, et al. Transcriptome profiling from adipose tissue during a low-calorie diet reveals predictors of weight and glycemic outcomes in obese, nondiabetic subjects. Am. J. Clin. Nutr. 2017;106:736–746. doi: 10.3945/ajcn.117.156216.
    1. Geyer PE, et al. Proteomics reveals the effects of sustained weight loss on the human plasma proteome. Mol. Syst. Biol. 2016;12:901. doi: 10.15252/msb.20167357.
    1. Oberbach A, et al. Combined proteomic and metabolomic profiling of serum reveals association of the complement system with obesity and identifies novel markers of body fat mass changes. J. Proteome Res. 2011;10:4769–4788. doi: 10.1021/pr2005555.
    1. Shah RV, et al. Proteins Altered by Surgical Weight Loss Highlight Biomarkers of Insulin Resistance in the Community. Arterioscler. Thromb. Vasc. Biol. 2019;39:107–115. doi: 10.1161/atvbaha.118.311928.
    1. Oller Moreno, S. et al. The differential plasma proteome of obese and overweight individuals undergoing a nutritional weight loss and maintenance intervention. Proteomics Clin Appl12, 10.1002/prca.201600150 (2018).
    1. Cominetti O, et al. Obesity shows preserved plasma proteome in large independent clinical cohorts. Sci. Rep. 2018;8:16981. doi: 10.1038/s41598-018-35321-7.
    1. Carayol J, et al. Protein quantitative trait locus study in obesity during weight-loss identifies a leptin regulator. Nat. Commun. 2017;8:2084. doi: 10.1038/s41467-017-02182-z.
    1. Bruderer R, et al. Analysis of 1508 Plasma Samples by Capillary-Flow Data-Independent Acquisition Profiles Proteomics of Weight Loss and Maintenance. Mol. Cell Proteom. 2019;18:1242–1254. doi: 10.1074/mcp.RA118.001288.
    1. Thrush AB, et al. Diet-resistant obesity is characterized by a distinct plasma proteomic signature and impaired muscle fiber metabolism. Int. J. Obes. 2018;42:353–362. doi: 10.1038/ijo.2017.286.
    1. Gardner CD, et al. Effect of Low-Fat vs Low-Carbohydrate Diet on 12-Month Weight Loss in Overweight Adults and the Association With Genotype Pattern or Insulin Secretion: The DIETFITS Randomized Clinical Trial. Jama. 2018;319:667–679. doi: 10.1001/jama.2018.0245.
    1. Stanton MV, et al. DIETFITS study (diet intervention examining the factors interacting with treatment success) - Study design and methods. Contemp. Clin. Trials. 2017;53:151–161. doi: 10.1016/j.cct.2016.12.021.
    1. Lind L, et al. EpiHealth: a large population-based cohort study for investigation of gene-lifestyle interactions in the pathogenesis of common diseases. Eur. J. Epidemiol. 2013;28:189–197. doi: 10.1007/s10654-013-9787-x.
    1. Lundberg M, Eriksson A, Tran B, Assarsson E, Fredriksson S. Homogeneous antibody-based proximity extension assays provide sensitive and specific detection of low-abundant proteins in human blood. Nucleic Acids Res. 2011;39:e102. doi: 10.1093/nar/gkr424.
    1. Assarsson E, et al. Homogenous 96-plex PEA immunoassay exhibiting high sensitivity, specificity, and excellent scalability. PLoS One. 2014;9:e95192. doi: 10.1371/journal.pone.0095192.
    1. Carlsson AC, et al. Use of Proteomics To Investigate Kidney Function Decline over 5 Years. Clin. J. Am. Soc. Nephrol. 2017;12:1226–1235. doi: 10.2215/cjn.08780816.
    1. Ganna A, Lee D, Ingelsson E, Pawitan Y. Rediscovery rate estimation for assessing the validation of significant findings in high-throughput studies. Brief. Bioinform. 2015;16:563–575. doi: 10.1093/bib/bbu033.
    1. Ganna A, et al. Large-scale metabolomic profiling identifies novel biomarkers for incident coronary heart disease. PLoS Genet. 2014;10:e1004801. doi: 10.1371/journal.pgen.1004801.
    1. Montague CT, et al. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 1997;387:903–908. doi: 10.1038/43185.
    1. Garin-Shkolnik T, Rudich A, Hotamisligil GS, Rubinstein M. FABP4 attenuates PPARgamma and adipogenesis and is inversely correlated with PPARgamma in adipose tissues. Diabetes. 2014;63:900–911. doi: 10.2337/db13-0436.
    1. Toth S, et al. Elevated Circulating PCSK9 Concentrations Predict Subclinical Atherosclerotic Changes in Low Risk Obese and Non-Obese Patients. Cardiol. Ther. 2017;6:281–289. doi: 10.1007/s40119-017-0092-8.
    1. Filippatos TD, et al. Effects of increased body weight and short-term weight loss on serum PCSK9 levels - a prospective pilot study. Arch. Med. Sci. Atheroscler. Dis. 2017;2:e46–e51. doi: 10.5114/amsad.2017.70502.
    1. Abifadel M, et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat. Genet. 2003;34:154–156. doi: 10.1038/ng1161.
    1. Cohen JC, Boerwinkle E, Mosley TH, Jr., Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N. Engl. J. Med. 2006;354:1264–1272. doi: 10.1056/NEJMoa054013.
    1. Leander K, et al. Circulating Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Predicts Future Risk of Cardiovascular Events Independently of Established Risk Factors. Circulation. 2016;133:1230–1239. doi: 10.1161/circulationaha.115.018531.
    1. Shen Y, Chen X, He J, Liao D, Zu X. Axl inhibitors as novel cancer therapeutic agents. Life Sci. 2018;198:99–111. doi: 10.1016/j.lfs.2018.02.033.
    1. Bray GA, et al. The Science of Obesity Management: An Endocrine Society Scientific Statement. Endocr. Rev. 2018;39:79–132. doi: 10.1210/er.2017-00253.
    1. Irincheeva, I., Hager, J., Dayon, L. & Cominetti, O. Biomarkers for predicting degree of weight loss in male subjects. WO2016169806A1, (2016).
    1. Dixon, J. B., Dixon, A. F. & Raven, J. Methods for predicting weight loss success. US20110124121A1, (2010).
    1. Arner P, et al. FGF21 attenuates lipolysis in human adipocytes - a possible link to improved insulin sensitivity. FEBS Lett. 2008;582:1725–1730. doi: 10.1016/j.febslet.2008.04.038.
    1. Kharitonenkov A, et al. FGF-21 as a novel metabolic regulator. J. Clin. Invest. 2005;115:1627–1635. doi: 10.1172/jci23606.
    1. Berglund ED, et al. Fibroblast growth factor 21 controls glycemia via regulation of hepatic glucose flux and insulin sensitivity. Endocrinology. 2009;150:4084–4093. doi: 10.1210/en.2009-0221.
    1. Coskun T, et al. Fibroblast growth factor 21 corrects obesity in mice. Endocrinology. 2008;149:6018–6027. doi: 10.1210/en.2008-0816.
    1. Bobbert T, et al. Fibroblast growth factor 21 predicts the metabolic syndrome and type 2 diabetes in Caucasians. Diabetes Care. 2013;36:145–149. doi: 10.2337/dc12-0703.
    1. Zhang X, et al. Serum FGF21 levels are increased in obesity and are independently associated with the metabolic syndrome in humans. Diabetes. 2008;57:1246–1253. doi: 10.2337/db07-1476.
    1. Chu AY, et al. Novel locus including FGF21 is associated with dietary macronutrient intake. Hum. Mol. Genet. 2013;22:1895–1902. doi: 10.1093/hmg/ddt032.
    1. von Holstein-Rathlou S, et al. FGF21 Mediates Endocrine Control of Simple Sugar Intake and Sweet Taste Preference by the Liver. Cell Metab. 2016;23:335–343. doi: 10.1016/j.cmet.2015.12.003.
    1. Kleinert M, et al. Quantitative proteomic characterization of cellular pathways associated with altered insulin sensitivity in skeletal muscle following high-fat diet feeding and exercise training. Sci. Rep. 2018;8:10723. doi: 10.1038/s41598-018-28540-5.

Source: PubMed

3
Abonnere