High-throughput mediation analysis of human proteome and metabolome identifies mediators of post-bariatric surgical diabetes control

Jonathan M Dreyfuss, Yixing Yuchi, Xuehong Dong, Vissarion Efthymiou, Hui Pan, Donald C Simonson, Ashley Vernon, Florencia Halperin, Pratik Aryal, Anish Konkar, Yinong Sebastian, Brandon W Higgs, Joseph Grimsby, Cristina M Rondinone, Simon Kasif, Barbara B Kahn, Kathleen Foster, Randy Seeley, Allison Goldfine, Vera Djordjilović, Mary Elizabeth Patti, Jonathan M Dreyfuss, Yixing Yuchi, Xuehong Dong, Vissarion Efthymiou, Hui Pan, Donald C Simonson, Ashley Vernon, Florencia Halperin, Pratik Aryal, Anish Konkar, Yinong Sebastian, Brandon W Higgs, Joseph Grimsby, Cristina M Rondinone, Simon Kasif, Barbara B Kahn, Kathleen Foster, Randy Seeley, Allison Goldfine, Vera Djordjilović, Mary Elizabeth Patti

Abstract

To improve the power of mediation in high-throughput studies, here we introduce High-throughput mediation analysis (Hitman), which accounts for direction of mediation and applies empirical Bayesian linear modeling. We apply Hitman in a retrospective, exploratory analysis of the SLIMM-T2D clinical trial in which participants with type 2 diabetes were randomized to Roux-en-Y gastric bypass (RYGB) or nonsurgical diabetes/weight management, and fasting plasma proteome and metabolome were assayed up to 3 years. RYGB caused greater improvement in HbA1c, which was mediated by growth hormone receptor (GHR). GHR's mediation is more significant than clinical mediators, including BMI. GHR decreases at 3 months postoperatively alongside increased insulin-like growth factor binding proteins IGFBP1/BP2; plasma GH increased at 1 year. Experimental validation indicates (1) hepatic GHR expression decreases in post-bariatric rats; (2) GHR knockdown in primary hepatocytes decreases gluconeogenic gene expression and glucose production. Thus, RYGB may induce resistance to diabetogenic effects of GH signaling.Trial Registration: Clinicaltrials.gov NCT01073020.

Conflict of interest statement

For the parent study (SLIMM-T2D) from which samples were derived for analysis in the current study, D.S., A.V., F.H., and A.B.G. received unrestricted funding from the Herbert Graetz Fund and from Covidien, to support surgical procedures for research participants with BMI 30-35 (not covered by insurance). Some supplies for the SLIMM-T2D clinical study were received from Lifescan, a division of Johnson and Johnson, Nestle Inc, and Novo Nordisk. M.E.P. received unrestricted investigator-initiated research grant funding from Medimmune to support the present work, and funding to support assay costs from SomaLogic. ABG served on advisory boards for Baranova, and Kowa. DCS received funds from PCORI and is a stock/shareholder of GI Windows. A.K., B.H., C.M.R., J.G., and Y.S. were employees of Medimmune when the work was initiated. Y.Y. is now employed at Vertex, F.H. at Form Health, A.K. at Sanofi, B.H. at Immunocore, J.G. at AstraZeneca, and A.B.G. at Novartis Institutes of Biomedical Research. All other authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. Graphical overview shows study flow…
Fig. 1. Graphical overview shows study flow including clinical study schematic (top) and analysis (bottom).
Differential analyte and pathway analysis (bottom left) identified a network composed of top analytes from Valine, Leucine, and Isoleucine Degradation and Beta-Alanine Metabolism. Mediation analysis (bottom right) identified GHR as mediator of decrease in HbA1c. Triangles denote measured proteins or metabolites, while circles denote proteins or metabolites in the pathway not measured; symbols are colored according to between-group Z scores, using heat map indicated, with the orientation of triangle also indicating up- or downregulation, respectively. Source data are provided as a Source Data file.
Fig. 2. Differentially abundant analytes with time.
Fig. 2. Differentially abundant analytes with time.
Heatmaps of a proteins and b metabolites. Heatmap of log2(RYGB/DWM) at all time points (post-baseline log2 abundance values are baseline-corrected) for analytes that are differentially abundant at any time point (FDR < 0.15 and fold-change absolute value > 1.5). The range of colors show log2(RYGB/DWM) from −1.6 to 1.6, which corresponds to fold changes from −3 to 3. Analytes with a larger absolute value of fold-change are shown as only having fold change absolute value of 3, so that weaker fold changes are easily observed. Source data are provided as a Source Data file.
Fig. 3. Phospholipid biosynthesis and other top-ranking…
Fig. 3. Phospholipid biosynthesis and other top-ranking differential pathways.
a Top-ranking differential pathways. Graph illustrates –log10(p-values) and false discovery rates (FDRs), with bars colored by FDR, from Limma Roast’s Mixed statistic (which is one-sided) adjusted with FDRs. b Phospholipid biosynthesis network. Nodes are colored by between-group z-score, whereas unmeasured nodes are colored dark gray. Orientation of triangle also indicates the directionality of regulation. Connections are from the Pathway Commons network. Source data are provided as a Source Data file.
Fig. 4. Branched chain amino acids (BCAA)…
Fig. 4. Branched chain amino acids (BCAA) and downstream metabolites.
a Schematic of BCAA metabolic pathway showing measured analytes. KIC, KMV, and KIV indicate the branched chain ketoacids ketoisocaproate, ketomethylvalerate, and ketoisovalerate, respectively. b Relative abundance of measured analytes, aligned to position in pathway in (a). Data were analyzed by two-sided moderated t-tests; post-baseline time points were analyzed using change from baseline. Data are reported as mean ± SEM on the log2 scale. Analysis was derived from samples from independent human participants. RYGB at 0, 3, 12, and 36 months: n = 19, 18, 19, and 13; DWM: n = 19, 18, 16, 9. Nominal p-values are as follows: Leucine: 3 mo **= 0.001, 12 mo **= 0.0041, 36 mo *= 0.03; Isoleucine: 12 mo *= 0.017, 36 mo *= 0.036; Valine: 3 mo **= 0.0019, 12 mo **= 0.0013, 36 mo *= 0.016; KIC: 3 mo ***= 0.00016; KIV: 3 mo: ***= 0.00026, 12 mo: **= 0.0045. 3-OH-isobutyrate: 3 mo: #<0.0001; isovalerylcarnitine: 3 mo: #<0.0001, 12 mo: **= 0.002; 2-methylbutyrylcarnitine: 0 mo: *= 0.025; propionylcarnitine: 3 mo: #<0.0001, 12 mo: *= 0.016. isobutyrylcarnitine: 0 mo: *= 0.024. Source data and FDRs are available in Supplementary Data 2.
Fig. 5. Beta-alanine metabolism.
Fig. 5. Beta-alanine metabolism.
a Network nodes are colored by between-group z-score, whereas unmeasured nodes are colored gray. Orientation of triangle also indicates the directionality of regulation. Connections are from the Pathway Commons network. b Log2 abundance of histidine measured by metabolomics in samples from independent human participants at baseline, 3, 12, and 36 months from 19, 18, 16, and 9 DWM and 19, 18, 19, and 13 RYGB participants. c Log2 abundance of CNDP1 measured by SOMAscan, measured in samples from independent human participants at baseline, 3, 12, 24, and 36 months from 19, 19, 16, 10, and 9 DWM and 19, 19,19, 15, and 14 RYGB participants. d CNDP1 plasma levels measured by ELISA at baseline, 3, 12, 24, and 36 months from 10, 10, 10, 6, and 4 DWM participants and 10, 10, 9, 5, and 6 RYGB participants, respectively. Data were analyzed by two-sided moderated t-tests; post-baseline time points were analyzed using change from baseline. Data in bd are reported as mean ± SEM. Nominal p-values are as follows: b histidine: 3 mo: #<0.0001, 36 mo: *=0.019; c CNDP1 (SOMAscan): 3 mo: #<0.0001, 12 mo: ***=0.00014; 24 mo: **=0.0054; 36 mo: **=0.0067; d CNDP1 ELISA: 3 mo: **=0.0036. Supplementary Data 2 includes FDRs and source data for (b/c); source data for d is provided as a Source Data file.
Fig. 6. GHR is a consistent mediator…
Fig. 6. GHR is a consistent mediator of change in HbA1c.
Retinol is an inconsistent mediator (i.e., it suppresses or inhibits the causal effect; left side) whereas GHR is a consistent mediator of HbA1c change at 1 year (right side). aX-axis represents log2 abundance change from baseline to the 3 month time point, and Y-axis represents HbA1c change from baseline at 1 year. b Shows putatively causal links with arrows based on (A) and direction of change with triangles pointed up (increase) or down (decrease). Source data are provided as a Source Data file.
Fig. 7. GHR is reduced after bariatric…
Fig. 7. GHR is reduced after bariatric surgery in humans and rodents and regulates hepatic glucose metabolism.
a GHR in plasma is reduced after RYGB in humans, measured by SOMAscan in samples from independent human participants at baseline, 3, 12, 24, and 36 months from 19, 19, 16, 10, and 9 DWM and 19, 19,19, 15, and 14 RYGB participants. bGhr expression in liver, assessed by qRT-PCR, from diet-induced obese rats, 8 weeks post-VSG bariatric surgery (VSG) or sham controls (Sham) (n = 13 VSG, n = 15 sham). cg Expression of GH signaling/axis genes, assessed by qRT-PCR, in cells treated with siRNA targeting Ghr (si-Ghr) or non-targeting scrambled control siRNA (NT-ctrl) (n = 6 per group). c Ghr, d Igf-1, and e Socs1/2 in mouse primary hepatocytes treated with siRNA-Ghr or NT-control. f Glucose production in response to insulin, cAMP, and insulin/cAMP after si-Ghr or NT-ctrl. g Expression of gluconeogenic genes G6pc and Pepck in response to si-Ghr or NT-ctrl. Data are reported as mean ± SEM. For a the p-values are nominal, using a two-sided T-test; FDRs are reported in Supplementary Data 2. For be and g two-sided t-tests were applied; for f two-way ANOVA with Bonferroni’s multiple comparison test was applied in GraphPad Prism. P values are as follows: a: GHR: 3 mo: ***= 0.00099; 12 mo: #<0.0001, 24 mo: **= 0.0024, 36 mo: **= 0.0013. b: *= 0.0155, c: # <0.0001, d: # <0.0001; e: Socs1: *= 0.017, Socs2: ***= 0.0002; f: basal vs. insulin in NT control: *= 0.0322; basal vs. cAMP in NT control: ***= 0.0004; NT control vs. siGHR in cAMP + insulin: *= 0.0328. g: G6pc and Pepck both #<0.0001. Source data for a is provided in Supplementary Data 2. Source data for panels bg are provided in the Source Data file. For expression analysis, two independent experiments were performed, while glucose production data represent a single experiment.

References

    1. Schauer PR, et al. Bariatric surgery versus intensive medical therapy for diabetes—5-year outcomes. N. Engl. J. Med. 2017;376:641–651.
    1. Mingrone G, et al. Bariatric-metabolic surgery versus conventional medical treatment in obese patients with type 2 diabetes: 5 year follow-up of an open-label, single-centre, randomised controlled trial. Lancet. 2015;386:964–973.
    1. Courcoulas AP, et al. Three-year outcomes of bariatric surgery vs lifestyle intervention for type 2 diabetes mellitus treatment: a randomized clinical trial. JAMA Surg. 2015;150:931–940.
    1. Cummings DE, et al. Gastric bypass surgery vs intensive lifestyle and medical intervention for type 2 diabetes: the CROSSROADS randomised controlled trial. Diabetologia. 2016;59:945–953.
    1. Simonson D, Halperin F, Foster K, Vernon A, Goldfine A. Clinical and patient-centered outcomes in obese patients with type 2 diabetes 3 years after randomization to Roux-en-Y gastric bypass surgery versus intensive lifestyle management: the SLIMM-T2D study. Diabetes Care. 2018;41:670–679.
    1. Jorgensen NB, et al. Acute and long-term effects of Roux-en-Y gastric bypass on glucose metabolism in subjects with type 2 diabetes and normal glucose tolerance. Am. J. Physiol. Endocrinol. Metab. 2012;303:E122–E131.
    1. Ramracheya RD, et al. PYY-dependent restoration of impaired insulin and glucagon secretion in type 2 diabetes following Roux-En-Y gastric bypass surgery. Cell Rep. 2016;15:944–950.
    1. Salehi M, Prigeon RL, D’Alessio DA. Gastric bypass surgery enhances glucagon-like Peptide 1-stimulated postprandial insulin secretion in humans. Diabetes. 2011;60:2308–2314.
    1. Laferrere B, et al. Differential metabolic impact of gastric bypass surgery versus dietary intervention in obese diabetic subjects despite identical weight loss. Sci. Transl. Med. 2011;3:80re82.
    1. Arora T, et al. Roux-en-Y gastric bypass surgery induces early plasma metabolomic and lipidomic alterations in humans associated with diabetes remission. PLoS One. 2015;10:e0126401.
    1. Dutia R, et al. Temporal changes in bile acid levels and 12alpha-hydroxylation after Roux-en-Y gastric bypass surgery in type 2 diabetes. Int. J. Obes. 2015;39:806–813.
    1. Patti, M. E. et al. Serum bile acids are higher in humans with prior gastric bypass: potential contribution to improved glucose and lipid metabolism. Obesity17, 1671−1677 (2009).
    1. Ryan KK, et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014;509:183–188.
    1. Liu H, Hu C, Zhang X, Jia W. Role of gut microbiota, bile acids and their cross-talk in the effects of bariatric surgery on obesity and type 2 diabetes. J. Diabetes Investig. 2018;9:13–20.
    1. DePaoli AM, et al. FGF19 analog as a surgical factor mimetic that contributes to metabolic effects beyond glucose homeostasis. Diabetes. 2019;68:1315–1328.
    1. Saeidi N, et al. Reprogramming of intestinal glucose metabolism and glycemic control in rats after gastric bypass. Science. 2013;341:406–410.
    1. Magouliotis D, Tasiopoulou V, Sioka E, Chatedaki C, Zacharoulis D. Impact of bariatric surgery on metabolic and gut microbiota profile: a systematic review and meta-analysis. Obes. Surg. 2017;27:1345–1357.
    1. Ilhan ZE, et al. Distinctive microbiomes and metabolites linked with weight loss after gastric bypass, but not gastric banding. ISME J. 2017;11:2047–2058.
    1. Aron-Wisnewsky, J. et al. Major microbiota dysbiosis in severe obesity: fate after bariatric surgery. Gut68, 70−82 (2018).
    1. Mokadem M, Zechner JF, Margolskee RF, Drucker DJ, Aguirre V. Effects of Roux-en-Y gastric bypass on energy and glucose homeostasis are preserved in two mouse models of functional glucagon-like peptide-1 deficiency. Mol. Metab. 2014;3:191–201.
    1. Ye J, et al. GLP-1 receptor signaling is not required for reduced body weight after RYGB in rodents. Am. J. Physiol. Regulatory, Integr. Comp. Physiol. 2014;306:R352–R362.
    1. Barfield R, et al. Testing for the indirect effect under the null for genome-wide mediation analyses. Genet. Epidemiol. 2017;41:824–833.
    1. MacKinnon DP, Lockwood CM, Hoffman JM, West SG, Sheets V. A comparison of methods to test mediation and other intervening variable effects. Psychol. Methods. 2002;7:83.
    1. Fritz MS, Mackinnon DP. Required sample size to detect the mediated effect. Psychol. Sci. 2007;18:233–239.
    1. Huang Y-T. Joint significance tests for mediation effects of socioeconomic adversity on adiposity via epigenetics. Ann. Appl. Stat. 2018;12:1535–1557.
    1. Tingley, D., Yamamoto, T., Hirose, K., Keele, L. & Imai, K. mediation: r package for causal mediation analysis. J. Stat. Softw.10.18637/jss.v059.i05 (2014).
    1. Al-Regaiey K, et al. Effects of gastric sleeve surgery on the serum levels of GH, IGF-1 and IGF-binding protein 2 in healthy obese patients. BMC Gastroenterol. 2020;20:199.
    1. Ceccarini G, et al. Serum IGF-binding protein 2 (IGFBP-2) concentrations change early after gastric bypass bariatric surgery revealing a possible marker of leptin sensitivity in obese subjects. Endocrine. 2019;65:86–93.
    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.
    1. Hedbacker K, et al. Antidiabetic effects of IGFBP2, a leptin-regulated gene. Cell Metab. 2010;11:11–22.
    1. Jewison T, et al. SMPDB 2.0: big improvements to the small molecule pathway database. Nucleic Acids Res. 2014;42:D478–D484.
    1. Rhodes CJ, et al. Plasma proteome analysis in patients with pulmonary arterial hypertension: an observational cohort study. Lancet Respir. Med. 2017;5:717–726.
    1. Yang Q, et al. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature. 2005;436:356–362.
    1. Mackinnon DP, Lockwood CM, Williams J. Confidence limits for the indirect effect: distribution of the product and resampling methods. Multivar. Behav. Res. 2004;39:99.
    1. Ritchie M, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:gkv007–e047.
    1. Barfield R, et al. Testing for the indirect effect under the null for genome‐wide mediation analyses. Genet Epidemiol. 2017;41:824–833.
    1. Law C, Chen Y, Shi W, Smyth G. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15:R29.
    1. Consortium GT. The genotype-tissue expression (GTEx) project. Nat. Genet. 2013;45:580–585.
    1. Leek JT, Storey JD. Capturing heterogeneity in gene expression studies by surrogate variable analysis. PLoS Genet. 2007;3:1724–1735.
    1. Oliva, M. et al. The impact of sex on gene expression across human tissues. Science10.1126/science.aba3066 (2020).
    1. Djordjilović V, et al. Global test for high-dimensional mediation: testing groups of potential mediators. Stat. Med. 2019;38:3346–3360.
    1. Huang YT, Pan WC. Hypothesis test of mediation effect in causal mediation model with high‐dimensional continuous mediators. Biometrics. 2015;72:402–413.
    1. Wu D, Smyth G. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res. 2012;40:e133–e133.
    1. Yoshino M, et al. Effects of diet versus gastric bypass on metabolic function in diabetes. N. Engl. J. Med. 2020;383:721–732.
    1. Aguiar-Oliveira MH, Bartke A. Growth hormone deficiency: health and longevity. Endocr. Rev. 2019;40:575–601.
    1. Pedersen MH, et al. Substrate metabolism and insulin sensitivity during fasting in obese human subjects: impact of GH blockade. J. Clin. Endocrinol. Metab. 2017;102:1340–1349.
    1. Ben-Zvi, D. et al. Time-dependent molecular responses differ between gastric bypass and dieting but are conserved across species. Cell Metabolism28, 310−323 (2018).
    1. Illan-Gomez F, et al. Obesity and inflammation: change in adiponectin, C-reactive protein, tumour necrosis factor-alpha and interleukin-6 after bariatric surgery. Obes. Surg. 2012;22:950–955.
    1. Wolf RM, et al. Cytokine, chemokine, and cytokine receptor changes are associated with metabolic improvements after bariatric surgery. J. Clin. Endocrinol. Metab. 2019;104:947–956.
    1. Mulla CM, et al. Plasma FGF-19 levels are increased in patients with post-bariatric hypoglycemia. Obes. Surg. 2019;29:2092–2099.
    1. Lips M, et al. Roux-en-Y gastric bypass surgery, but not calorie restriction, reduces plasma branched-chain amino acids in obese women independent of weight loss or the presence of type 2 diabetes. Diabetes Care. 2014;37:3150–3156.
    1. Wijayatunga, N. et al. Roux-en-Y gastric bypass surgery alters serum metabolites and fatty acids in patients with morbid obesity. Diabetes/Metab. Res. Rev.10.1002/dmrr.3045 (2018).
    1. Wang T, et al. Metabolite profiles and the risk of developing diabetes. Nat. Med. 2011;17:448–453.
    1. Newgard CB, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9:311–326.
    1. White PJ, et al. Branched-chain amino acid restriction in Zucker-fatty rats improves muscle insulin sensitivity by enhancing efficiency of fatty acid oxidation and acyl-glycine export. Mol. Metab. 2016;5:538–551.
    1. Jang C, et al. A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance. Nat. Med. 2016;22:421–426.
    1. Viegas CM, et al. Evidence that 3-hydroxyisobutyric acid inhibits key enzymes of energy metabolism in cerebral cortex of young rats. Int. J. Dev. Neurosci.: Off. J. Int. Soc. Dev. Neurosci. 2008;26:293–299.
    1. Juiz-Valina, P. et al. Altered GH-IGF-1 axis in severe obese subjects is reversed after bariatric surgery-induced weight loss and related with low-grade chronic inflammation. J. Clin. Med.10.3390/jcm9082614 (2020).
    1. Kopchick JJ, Berryman DE, Puri V, Lee KY, Jorgensen JOL. The effects of growth hormone on adipose tissue: old observations, new mechanisms. Nat. Rev. Endocrinol. 2020;16:135–146.
    1. Melmed S. Pathogenesis and diagnosis of growth hormone deficiency in adults. N. Engl. J. Med. 2019;380:2551–2562.
    1. Dominici FP, Arostegui Diaz G, Bartke A, Kopchick JJ, Turyn D. Compensatory alterations of insulin signal transduction in liver of growth hormone receptor knockout mice. J. Endocrinol. 2000;166:579–590.
    1. Fang F, Shi X, Brown MS, Goldstein JL, Liang G. Growth hormone acts on liver to stimulate autophagy, support glucose production, and preserve blood glucose in chronically starved mice. Proc. Natl Acad. Sci. USA. 2019;116:7449–7454.
    1. Sjostrom L, et al. Association of bariatric surgery with long-term remission of type 2 diabetes and with microvascular and macrovascular complications. Jama. 2014;311:2297–2304.
    1. Arterburn D. Microvascular outcomes in patients with diabetes after bariatric surgery. Ann. Intern. Med. 2019;170:507–508.
    1. Fisher DP, et al. Association between bariatric surgery and macrovascular disease outcomes in patients with type 2 diabetes and severe obesity. Jama. 2018;320:1570–1582.
    1. Aminian, A. et al. Association of metabolic surgery with major adverse cardiovascular outcomes in patients with type 2 diabetes and obesity. Jama10.1001/jama.2019.14231 (2019).
    1. Zhang H, et al. Estimating and testing high-dimensional mediation effects in epigenetic studies. Bioinformatics. 2016;32:3150–3154.
    1. Chén OY, et al. High-dimensional multivariate mediation with application to neuroimaging data. Biostatistics. 2018;19:121–136.
    1. VanderWeele TJ, Vansteelandt S. Mediation analysis with multiple mediators. Epidemiol. Method. 2014;2:95–115.
    1. Bland JM, Altman DG. One and two sided tests of significance. BMJ. 1994;309:248.
    1. Freedman LS. An analysis of the controversy over classical one-sided tests. Clin. Trials. 2008;5:635–640.
    1. Ludbrook J. Should we use one-sided or two-sided P values in tests of significance? Clin. Exp. Pharm. Physiol. 2013;40:357–361.
    1. Wolterbeek R, Enkin MW, Bland JM, Altman DG. One and two sided tests of significance statsitical hypothesis should be brought into line with clinial hypothesis. BMJ. 1994;309:873–874.
    1. Knottnerus JA, Bouter LM. The ethics of sample size: two-sided testing and one-sided thinking. J. Clin. Epidemiol. 2001;54:109–110.
    1. Cho H-C, Abe S. Is two-tailed testing for directional research hypotheses tests legitimate? J. Bus. Res. 2013;66:1261–1266.
    1. VanderWeele TJ. Mediation and mechanism. Eur. J. Epidemiol. 2009;24:217–224.
    1. Panosian J, et al. Physical activity in obese type 2 diabetes after gastric bypass or medical management. Am. J. Med. 2017;130:83–92.
    1. Gold L, Walker J, Wilcox S, Williams S. Advances in human proteomics at high scale with the SOMAscan proteomics platform. N. Biotechnol. 2012;29:543–549.
    1. Graham TE, et al. Retinol-binding protein 4 and insulin resistance in lean, obese, and diabetic subjects. N. Engl. J. Med. 2006;354:2552–2563.
    1. Gall WE, et al. alpha-hydroxybutyrate is an early biomarker of insulin resistance and glucose intolerance in a nondiabetic population. PLoS One. 2010;5:e10883.
    1. Ford L, et al. Precision of a clinical metabolomics profiling platform for use in the identification of inborn errors of metabolism. J. Appl. Lab. Med. 2020;5:342–356.
    1. Sumner LW, et al. Proposed minimum reporting standards for chemical analysis Chemical Analysis Working Group (CAWG) Metabolomics Standards Initiative (MSI) Metabolomics. 2007;3:211–221.
    1. Little, R. J. & Rubin, D. B. Statistical Analysis with Missing Data 793 (John Wiley & Sons, 2019).
    1. Wei, R. et al. Missing value imputation approach for mass spectrometry-based metabolomics data. Sci. Rep.10.1038/s41598-017-19120-0 (2018).
    1. Gentleman R, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5:1–16.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B. 1995;57:289–300.
    1. Wu D, et al. ROAST: rotation gene set tests for complex microarray experiments. Bioinformatics. 2010;26:2176–2182.
    1. Cerami E, et al. Pathway Commons, a web resource for biological pathway data. Nucleic Acids Res. 2011;39:D685–D690.
    1. Stefater MA, et al. Sleeve gastrectomy induces loss of weight and fat mass in obese rats, but does not affect leptin sensitivity. Gastroenterology. 2010;138:2426–2436.
    1. Jonathan M. Dreyfuss Analysis of omics from the SLIMM-T2D trial (v1.0.4). Zenodo10.5281/zenodo.5485746. (2021).

Source: PubMed

3
Suscribir