The plasma proteome identifies expected and novel proteins correlated with micronutrient status in undernourished Nepalese children

Robert N Cole, Ingo Ruczinski, Kerry Schulze, Parul Christian, Shelley Herbrich, Lee Wu, Lauren R Devine, Robert N O'Meally, Sudeep Shrestha, Tatiana N Boronina, James D Yager, John Groopman, Keith P West Jr, Robert N Cole, Ingo Ruczinski, Kerry Schulze, Parul Christian, Shelley Herbrich, Lee Wu, Lauren R Devine, Robert N O'Meally, Sudeep Shrestha, Tatiana N Boronina, James D Yager, John Groopman, Keith P West Jr

Abstract

Micronutrient deficiencies are common in undernourished societies yet remain inadequately assessed due to the complexity and costs of existing assays. A plasma proteomics-based approach holds promise in quantifying multiple nutrient:protein associations that reflect biological function and nutritional status. To validate this concept, in plasma samples of a cohort of 500 6- to 8-y-old Nepalese children, we estimated cross-sectional correlations between vitamins A (retinol), D (25-hydroxyvitamin D), and E (α-tocopherol), copper, and selenium, measured by conventional assays, and relative abundance of their major plasma-bound proteins, measured by quantitative proteomics using 8-plex iTRAQ mass tags. The prevalence of low-to-deficient status was 8.8% (<0.70 μmol/L) for retinol, 19.2% (<50 nmol/L) for 25-hydroxyvitamin D, 17.6% (<9.3 μmol/L) for α-tocopherol, 0% (<10 μmol/L) for copper, and 13.6% (<0.6 μmol/L) for selenium. We identified 4705 proteins, 982 in >50 children. Employing a linear mixed effects model, we observed the following correlations: retinol:retinol-binding protein 4 (r = 0.88), 25-hydroxyvitamin D:vitamin D-binding protein (r = 0.58), α-tocopherol:apolipoprotein C-III (r = 0.64), copper:ceruloplasmin (r = 0.65), and selenium:selenoprotein P isoform 1 (r = 0.79) (all P < 0.0001), passing a false discovery rate threshold of 1% (based on P value-derived q values). Individual proteins explained 34-77% (R(2)) of variation in their respective nutrient concentration. Adding second proteins to models raised R(2) to 48-79%, demonstrating a potential to explain additional variation in nutrient concentration by this strategy. Plasma proteomics can identify and quantify protein biomarkers of micronutrient status in undernourished children. The maternal micronutrient supplementation trial, from which data were derived as a follow-up activity, was registered at clinicaltrials.gov as NCT00115271.

Figures

FIGURE 1
FIGURE 1
Plasma retinol and RBP4 relative abundance distributions in Nepalese children 6–8 y of age (n = 500). (A) Histogram showing the frequency distribution of retinol concentrations: range = 0.30–2.11 μmol/L, 8.8% (n = 44) deficient (<0.70 μmol/L, dark gray), 45.6% (n = 228), marginal (0.70 to <1.05 μmol/L, medium gray), and 45.6% (n = 228) adequate (≥1.05 μmol/L, light gray) in status. (B) Plasma retinol by relative abundance of RBP4 by a traditional estimation method using a master plasma pool in one randomly assigned iTRAQ channel within each 8-plex experiment to normalize the protein distribution across iTRAQ runs. (C) Plasma retinol by relative abundance of RBP4 by an estimation method that relies on an LME model that combines abundance estimates from all 72 iTRAQ experiments (25). R2 values represent the proportion of variance in the nutrient explained by the fitted values of the nutrient-protein regression models. The P value in B is derived from testing the hypothesis of no association between the nutrient and protein abundance, whereas the P value in C is derived from testing the fixed effects slope for the protein abundance in the LME model. Shading of circles in B and C corresponds to bars. Horizontal lines indicate cutoffs for changes in micronutrient status. iTRAQ, isobaric tags for relative and absolute quantification; LME, linear mixed effects (model); RBP4, retinol binding protein isoform 4.
FIGURE 2
FIGURE 2
Plasma 25-hydroxyvitamin D and VDBP relative abundance distributions in Nepalese children 6–8 y of age (n = 500). (A) Frequency distribution of 25-hydroxyvitamin D concentrations: range, 18.6–173.5 nmol/L, 19.2% (n = 96) deficient (<50 nmol/L, dark gray), and 80.8% (n = 404, medium gray) adequate (≥50 nmol/L) in status. (B,C) Plasma 25-hydroxyvitamin D by relative abundance of VDBP by traditional master plasma pool normalization and LME-adjusted methods, respectively (see Fig. 1 for details). LME, linear mixed effects (model); VDBP, vitamin D binding protein; 25(OH)D, 25-hydroxyvitamin D.
FIGURE 3
FIGURE 3
Plasma α-tocopherol and Apo C-III relative abundance distributions in Nepalese children 6–8 y of age (n = 500). (A) Frequency distribution of α-tocopherol concentrations: range, 4.1–26.9 μmol/L, 17.6% (n = 88) deficient (<9.3 μmol/L, dark gray), 37.4% (n = 187) marginal (9.3 to <12 μmol/L, medium gray), and 45% (n = 225) adequate (≥12 μmol/L, light gray) in status. (B,C) Plasma α-tocopherol by relative abundance of Apo C-III by traditional master plasma pool normalization and LME-adjusted methods, respectively (see Fig. 1 for details). LME, linear mixed effects (model).
FIGURE 4
FIGURE 4
Plasma copper and Cp relative abundance distributions in Nepalese children 6–8 y of age (n = 494). (A) Plasma copper concentrations: range, 11.6–35.8 μmol/L, 100% were adequate (>10 μmol/L, gray). Six implausible values (4 <5 μmol/L, and 1 each at 62.3 μmol/L and 100.5 μmol/L) were removed from this analysis. (B,C) Plasma copper by relative abundance of Cp by traditional master plasma pool normalization and LME-adjusted methods, respectively (see Fig. 1 for details). Cp, ceruloplasmin; LME, linear mixed effects (model).
FIGURE 5
FIGURE 5
Plasma selenium and SEPP1 relative abundance distributions in Nepalese children 6–8 y of age (n = 499). (A) Plasma selenium concentrations: range, 0.4–2.1 μmol/L; 13.6% (n = 68) deficient (<0.6 μmol/L, dark gray) and 86.4% (n = 431) adequate (≥0.6 μmol/L, medium gray) in status. (B,C) Plasma selenium by relative abundance of SEPP1 by traditional master plasma pool normalization and LME-adjusted methods, respectively (see Fig. 1 for details). LME, linear mixed effects (model); SEPP1, selenoprotein P isoform 1.

References

    1. Arlappa N, Laxmaiah A, Balakrishna N, Harikumar R, Kodavanti MR. Gal Reddy Ch, Saradkumar S, Ravindranath M, Brahmam GN. Micronutrient deficiency disorders among the rural children of West Bengal, India. Ann Hum Biol. 2011;38:281–9.
    1. Pasricha SR, Shet AS, Black JF, Sudarshan H, Prashanth NS, Biggs BA. Vitamin B-12, folate, iron, and vitamin A concentrations in rural Indian children are associated with continued breastfeeding, complementary diet, and maternal nutrition. Am J Clin Nutr. 2011;94:1358–70.
    1. Jiang T, Christian P, Khatry SK, Wu L, West KP Jr. Micronutrient deficiencies in early pregnancy are common, concurrent, and vary by season among rural Nepali pregnant women. J Nutr. 2005;135:1106–12.
    1. West KP Jr, Pokhrel RP, Katz J, LeClerq SC, Khatry SK, Shrestha SR, Pradhan EK, Tielsch JM, Pandey MR, Sommer A. Efficacy of vitamin A in reducing preschool child mortality in Nepal. Lancet. 1991;338:67–71.
    1. Tielsch JM, Khatry SK, Stoltzfus RJ, Katz J, LeClerq SC, Adhikari R, Mullany LC, Black R, Shresta S. Effect of daily zinc supplementation on child mortality in southern Nepal: a community-based, cluster randomised, placebo-controlled trial. Lancet. 2007;370:1230–9.
    1. Christian P, Khatry SK, Katz J, Pradhan EK, LeClerq SC, Shrestha SR, Adhikari RK, Sommer A, West KP Jr. Effects of alternative maternal micronutrient supplements on low birth weight in rural Nepal. A double-masked randomized community trial. BMJ. 2003;326:571–6.
    1. Christian P, Murray-Kolb LE, Khatry SK, Katz J, Schaefer BA, Cole PM, LeClerq SC, Tielsch JM. Prenatal micronutrient supplementation and intellectual and motor function in early school-aged children in Nepal. JAMA. 2010;304:2716–23.
    1. Christian P, Stewart CP, LeClerq SC, Wu L, Katz J, West KP Jr, Khatry SK. Antenatal and postnatal iron supplementation and childhood mortality in rural Nepal: a prospective follow-up in a randomized, controlled community trial. Am J Epidemiol. 2009;170:1127–36.
    1. Raiten DJ, Namasté S, Brabin B, Combs G Jr, L'Abbe MR, Wasantwisut E, Darnton-Hill I. Executive summary: biomarkers of nutrition for development: building a consensus. Am J Clin Nutr. 2011;94:S633–50.
    1. Erhardt JG, Estes JE, Pfeiffer CM, Biesalski HK, Craft NE. Combined measurement of ferritin, soluble transferrin receptor, retinol binding protein, and C-reactive protein by an inexpensive, sensitive, and simple sandwich enzyme-linked immunosorbent assay technique. J Nutr. 2004;134:3127–32.
    1. Bechir M, Schelling E, Kraemer K, Schweigert F, Bonfoh B, Crump L, Tanner M, Zinsstag J. Retinol assessment among women and children in Sahelian mobile pastoralists. EcoHealth. 2012;9:113–21.
    1. Pierce A, Unwin RD, Evans CA, Griffiths S, Carney L, Zhang L, Jaworska E, Lee C-F, Blinco D, Okoniewski MJ, et al. . Eight-channel iTRAQ enables comparison of the activity of six leukemogenic tyrosine kinases. Mol Cell Proteomics. 2008;7:853–63.
    1. Farrah T, Deitsch EW, Omenn GS, Campbell DS, Sun Z, Bletz JA, Mallick P, Katz JE, Malmstrom J, Ossola R, et al. . A high-confidence human plasma proteome reference set with estimated concentrations in PeptideAtlas. Mol Cell Proteomics. 2011;10:M110.006353.
    1. Barnes S, Kim H. Nutriproteomics: identifying the molecular targets of nutritive and non-nutritive components of the diet. J Biochem Mol Biol. 2004;37:59–74.
    1. Schweigert FJ. Nutritional proteomics: methods and concepts for research in nutritional science. Ann Nutr Metab. 2007;51:99–107.
    1. Zhang X, Yap Y, Wei D, Chen G, Chen F. Novel omics technologies in nutrition research. Biotechnol Adv. 2008;26:169–76.
    1. Sénéchal S, Kussmann M. Nutriproteomics: technologies and applications for identification and quantification of biomarkers and ingredients. Proc Nutr Soc. 2011;70:351–64.
    1. Quadro L, Hamburger L, Colantuoni V, Gottesman ME, Blaner WS. Understanding the physiological role of retinol-binding protein in vitamin A metabolism using transgenic and knockout mouse models. Mol Aspects Med. 2003;24:421–30.
    1. Christakos S, Ajibade DV, Dhawan P, Fechner AJ, Mady LJ. Vitamin D: metabolism. Endocrinol Metab Clin North Am. 2010;39:243–53.
    1. Borel P, Moussa M, Reboul E, Lyan B, Defoort C, Vincent-Baudry S, Maillot M, Gastaldi M, Darmon M, Portugal H, et al. . Human fasting plasma concentrations of vitamin E and carotenoids, and their association with genetic variants in apo C–III, cholesteryl ester transfer protein, hepatic lipase, intestinal fatty acid binding protein and microsomal triacylglycerol transfer protein. Br J Nutr. 2009;101:680–7.
    1. Danzeisen R, Araya M, Harrison B, Keen C, Solioz M, Thiele D, McArdle HJ. How reliable and robust are current biomarkers for copper status? Br J Nutr. 2007;98:676–83.
    1. Burk RF, Hill KE. Selenoprotein P-expression, functions, and roles in mammals. Biochim Biophys Acta. 2009;1790:1441–7.
    1. Stewart CP, Christian P, Schulze KJ, LeClerq SC, West KP Jr, Khatry SK. Antenatal micronutrient supplementation reduces metabolic syndrome in 6- to 8-year-old children in rural Nepal. J Nutr. 2009;139:1575–81.
    1. WHO Growth reference data for 5–19 years. Geneva: WHO; 2007. [cited 2012 Oct 11]. Available from: .
    1. Herbrich SM, Cole RN, West KP Jr, Schulze K, Yager JD, Groopman JD, Christian P, Wu L, O'Meally RN, May DH, et al. . Statistical inference from multiple iTRAQ experiments without using common reference standards. J Proteome Res. 2013;12:594–604.
    1. Robinson GK. That BLUP is a good thing: the estimation of random effects. Stat Sci. 1991;6:15–32.
    1. The Comprehensive R Archive Network Available from: .
    1. West KP., Jr Extent of vitamin A deficiency among preschool children and women of reproductive age. J Nutr. 2002;132:S2857–66.
    1. Greer FR. 25-hydroxyitamin D: functional outcomes in infants and young children. Am J Clin Nutr. 2008;88:S529–33.
    1. Institute of Medicine Dietary reference intakes for vitamin C, vitamin E, selenium, and carotenoids. Washington: National Academy Press; 2000. p. 186–283.
    1. Harvey LJ, Ashton K, Hooper L, Casgrain A, Fairweather-Tait SJ. Methods of assessment of copper status in humans: a systematic review. Am J Clin Nutr. 2009;89:S2009–24.
    1. Ashton K, Hooper L, Harvey LJ, Hurst R, Casgrain A, Fairweather-Tait SJ. Methods of assessment of selenium status in humans: a systematic review. Am J Clin Nutr. 2009;89:S2025–39.
    1. Storey JD. A direct approach to false discovery rates. JRSS-B. 2002;64:479–98.
    1. de Pee S, Dary O. Biochemical indicators of vitamin A deficiency: serum retinol and serum retinol binding protein. J Nutr. 2002;132:S2895–901.
    1. Yang Q, Graham TE, Mody N, Preitner F, Peroni OD, Zabolotny JM, Kotani K, Quadro L, Kahn BB. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature. 2005;436:356–62.
    1. Mayilyan KR. Complement genetics, deficiencies, and disease associations. Protein Cell. 2012;3:487–96.
    1. Speeckaert M, Huang G, Delanghe JR, Taes YEC. Biological and clinical aspects of the vitamin D binding protein (Gc-globulin) and its polymorphism. Clin Chim Acta. 2006;372:33–42.
    1. Takamatsu H, Kumanogoh A. Diverse roles for semaphorin-plexin signaling in the immune system. Trends Immunol. 2012;33:127–35.
    1. Kang S, Kumanogoh A. Semaphorins in bone development, homeostasis, and disease. Semin Cell Dev Biol. 2013;24:163–71.
    1. Sutton AL, Zhang X, Dowd DR, Kharode YP, Komm BS, Macdonald PN. Semaphorin 3B is a 1,25-dihydroxyvitamin D3-induced gene in osteoclasts that promotes osteoclastogenesis and induces osteopenia in mice. Mol Endocrinol. 2008;22:1370–81.
    1. Fischer PR, Thacher TD, Pettifor JM. Pediatric vitamin D and calcium nutrition in developing countries. Rev Endocr Metab Disord. 2008;9:181–92.
    1. Holl EK, O'Connor BP, Holl TM, Roney KE, Zimmermann AG, Jha S, Kelsoe G, Ting JP. Plexin-D1 is a novel regulator of germinal centers and humoral immune responses. J Immunol. 2011;186:5603–11.
    1. Peelen E, Knippenberg S, Muris AH, Thewissen M, Smolders J, Tervaert JW, Hupperts R, Damoiseaux J. Effects of vitamin D on the peripheral adaptive immune system: a review. Autoimmun Rev. 2011;10:733–43.
    1. Serini G, Valdembri D, Zanivan S, Morterra G, Burkhardt C, Caccavari F, Zammataro L, Primo L, Tamagnone L. Class 3 semaphorins control vascular morphogenesis by inhibiting integrin function. Nature. 2003;424:391–7.
    1. Gu C, Yoshida Y, Livet J, Reimert DV, Mann F, Merte J, Henderson CE, Jessell TM, Kolodkin AL, Ginty DD. Semaphorin 3E and plexin-D1 control vascular pattern independently of neuropilins. Science. 2005;307:265–8.
    1. Garcia LA, Ferrini MG, Norris KC, Artaza JN. 1,25(OH)2vitamin D3 enhances myogenic differentiation by modulating the expression of key angiogenic growth factors and angiogenic inhibitors in CC skeletal muscle cells. J Steroid Biochem Mol Biol. 2013;133:1–11.
    1. Morrissey PA, Kiely M. Vitamin E/physiology and health effects. In: Caballero B, Allen L, Prentice A eds. Encyclopedia of human nutrition. 2nd ed Amsterdam: Elsevier Academic Press; 2005. p. 389–98.
    1. Mendivil CO, Zheng C, Furtado J, Lel J, Sacks FM. Metabolism of VLDL and LDL containing apolipoprotein C–III and not other small apolipoproteins-R2. Arterioscler Thromb Vasc Biol. 2010;30:239.
    1. Saitoh O, Yoshihiro K. Biochemical and electrophysiological analyses of RGS8 function. Methods Enzymol. 2004;390:129–48.
    1. Miyamoto-Matsubara M, Saitoh O, Maruyama K, Aizaki Y, Saito Y. Regulation of melanin-concentrating hormone receptor 1 signaling by RGS8 with the receptor third intracellular loop. Cell Signal. 2008;20:2084–94.
    1. Harvey LJ, McArdle HJ. Biomarkers of copper status: a brief update. Br J Nutr. 2008;99:S10–3.
    1. Hellman NE, Gitlin JD. Ceruloplasmin metabolism and function. Annu Rev Nutr. 2002;22:439–58.
    1. Meyer LA, Durley AP, Prohaska JR, Harris ZL. Copper transport and metabolism are normal in aceruloplasminemic mice. J Biol Chem. 2001;276:36857–61.
    1. Tsai C-Y, Finley JC, Ali SS, Patel HH, Howell SB. Copper influx transporter 1 is required for FGF, PDGF and EGF-induced MAPK signaling. Biochem Pharmacol. 2012;84:1007–13.
    1. Yang JG, Hill KE, Burk RF. Dietary selenium intake controls rat plasma selenoprotein P concentration. J Nutr. 1989;119:1010–2.
    1. Burk RF, Hill KE, Motley AK, Austin LM, Norsworthy BK. Deletion of selenoprotein P upregulates urinary selenium excretion and depresses whole-body selenium content. Biochim Biophys Acta 2006;1760:1789–93.
    1. Deagen JT, Butler JA, Zachara BA, Whanger PD. Determination of the distribution of selenium between glutathione peroxidase, selenoprotein P, and albumin in plasma. Anal Biochem. 1993;208:176–81.
    1. Burk RF, Olsen GE, Winfrey VP, Hill KE, Yin D. Glutathione peroxidase-3 produced by the kidney binds to population basement membranes in the gastrointestinal tract and other tissues. Am J Physiol Gastrointest Liver Physiol. 2011;301:G32–8.
    1. Wang P, Tang H, Zhang H, Whiteaker J, Paulovich AG, McIntosh M. Normalization regarding non-random missing values in high-throughput mass spectrometry data. Pac Symp Biocomput. 2006;315–26.
    1. Rubin DB. Multiple imputation after 18+ years (with discussion). JASA. 1996;91:473–89.
    1. Little RJA, An H. Robust likelihood-based analysis of multivariate data with missing values. Statist Sinica. 2004;14:949–68.
    1. Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics. 2002;1:845–67.

Source: PubMed

3
Subscribe