Proteomics reveals the effects of sustained weight loss on the human plasma proteome

Philipp E Geyer, Nicolai J Wewer Albrechtsen, Stefka Tyanova, Niklas Grassl, Eva W Iepsen, Julie Lundgren, Sten Madsbad, Jens J Holst, Signe S Torekov, Matthias Mann, Philipp E Geyer, Nicolai J Wewer Albrechtsen, Stefka Tyanova, Niklas Grassl, Eva W Iepsen, Julie Lundgren, Sten Madsbad, Jens J Holst, Signe S Torekov, Matthias Mann

Abstract

Sustained weight loss is a preferred intervention in a wide range of metabolic conditions, but the effects on an individual's health state remain ill-defined. Here, we investigate the plasma proteomes of a cohort of 43 obese individuals that had undergone 8 weeks of 12% body weight loss followed by a year of weight maintenance. Using mass spectrometry-based plasma proteome profiling, we measured 1,294 plasma proteomes. Longitudinal monitoring of the cohort revealed individual-specific protein levels with wide-ranging effects of losing weight on the plasma proteome reflected in 93 significantly affected proteins. The adipocyte-secreted SERPINF1 and apolipoprotein APOF1 were most significantly regulated with fold changes of -16% and +37%, respectively (P < 10-13), and the entire apolipoprotein family showed characteristic differential regulation. Clinical laboratory parameters are reflected in the plasma proteome, and eight plasma proteins correlated better with insulin resistance than the known marker adiponectin. Nearly all study participants benefited from weight loss regarding a ten-protein inflammation panel defined from the proteomics data. We conclude that plasma proteome profiling broadly evaluates and monitors intervention in metabolic diseases.

Keywords: diabetes; mass spectrometry; metabolic syndrome; obesity; plasma proteome profiling.

© 2016 The Authors. Published under the terms of the CC BY 4.0 license.

Figures

Figure 1. Study design and the plasma…
Figure 1. Study design and the plasma proteome profile pipeline
  1. The study cohort consisted of 52 obese individuals, who lost on average 12% of their body mass during 8 weeks of calorie restriction (800 kcal/day). The acute weight loss was followed by a 52‐week weight maintenance period by 43 of the study participants with longitudinal blood sampling at the indicated time points.

  2. Quadruplicates of the samples and the establishment of a matching library resulted in 1,294 plasma proteomes, which were separately prepared by an automated liquid handling platform. The LC‐MS/MS data, which we analyzed by MaxQuant and Perseus, resulted in 319 individual plasma proteome profiles for 52 participants.

Figure EV1. MS ‐based measurements and sample…
Figure EV1. MS‐based measurements and sample quality
  1. Total MS measuring time leading to usable raw data or lost to HPLC issues, MS cleaning, or other problems. The 6% cleaning time for the MS instrument resulted from a single cleaning instance, which, however, subsequently led to a 5‐day downtime.

  2. Analysis of the reproducibility between samples by color‐coded pairwise comparison (Pearson correlation coefficients) of 1,276 samples (without the matching library). The high reproducibility within quadruplicates and the individuals can be seen by the red diagonal, indicating high correlation.

  3. Levels of four highly abundant erythrocyte‐specific proteins were used to assess the sample quality with regard to erythrocyte lysis. The red arrow indicates the only sample with strong erythrocyte lysis.

  4. Displaying the levels of fibrinogens allows the detection of (partial) blood coagulation. Five cases with decreased fibrinogen levels and therefore coagulation events are highlighted by red arrows.

Figure 2. Individual‐specific plasma protein levels
Figure 2. Individual‐specific plasma protein levels
  1. Coefficients of variation (CVs) for all proteins were calculated in all participants for the five longitudinal samples. Combination of these longitudinal CVs and fold differences allows estimation of how many proteins are individual‐specific.

  2. CVs plotted against the log2‐fold difference of one participant compared to the average label‐free quantitation (LFQ) intensity of the study cohort. Considering a fivefold difference and a CV below 30% yields the proteins in the blue boxes as specific for this participant.

  3. LFQ intensities of seven proteins plotted during weight maintenance (weeks 4–52) for all 43 individuals. These proteins are stable over time within individuals, but strongly vary between individuals. A2M: alpha‐2‐macroglobulin; PZP: pregnancy zone protein; IGJ: immunoglobulin J chain; LPA: apolipoprotein(a); FETUB: fetuin‐B; CRP: C‐reactive protein; GDI1/GDI2: Rab GDP dissociation inhibitor alpha/beta.

Figure 3. The effect of weight loss
Figure 3. The effect of weight loss
  1. Volcano plot of proteomes before (week −8) and directly after weight loss (week 0), with the x‐axis depicting the fold change in protein levels and the y‐axis the –log10t‐test P‐value of the quadruplicates.

  2. Changes in protein abundance are shown for each individual. LFQ intensities from before (yellow) and after weight loss (green) are connected by a line to highlight individual‐specific protein levels and their changes. The median increase or decrease for each protein over the population is indicated in red.

Figure 4. Long‐term effects of weight loss…
Figure 4. Long‐term effects of weight loss on the plasma proteome profile
Hierarchical clustering of Z‐scored median LFQ intensities for highly significant proteins (P < 0.0005) resulted in seven longitudinal weight regulated protein clusters. Scale bar for Z‐scored weight is displayed below the protein clusters. Insets show Z‐scores for proteins in different clusters as a function of time, color‐coded for the distance from the center. The highlighted protein names with the same color indicate functionally connected proteins (red: inflammatory markers; brown: serine protease inhibitors; orange: complement system; blue: apolipoproteins; purple: anti‐inflammatory‐acting proteins; green: steroid transport proteins).
Figure EV2. Proteins with significant long‐term effects…
Figure EV2. Proteins with significant long‐term effects due to weight loss
  1. Proteins with a significant increase in at least five out of six time points, comparing before and after weight loss.

  2. Proteins with a significant decrease over a long time period due to weight loss.

  3. Long‐term behavior of specific proteins. The means are plotted with SEM as error bars over time. Upregulated proteins are indicated in orange and downregulated in blue.

Figure 5. Plasma proteins and the longitudinal…
Figure 5. Plasma proteins and the longitudinal inflammation profile
  1. A

    Correlation of body mass index (BMI) with all quantified proteins in our dataset.

  2. B–F

    Correlation analysis of the indicated clinical parameter with plasma protein levels over time.

  3. G

    For each individual, Z‐scores were calculated for each of the proteins of the ten‐protein panel over the seven time points. The proteins were arranged in the indicated order and hierarchical clustering was performed on the level of the different individuals, resulting in a longitudinal inflammation profile.

  4. H

    Dot plots for the ten‐protein panel in the same order as in (G) for the central cluster. The black line indicates the median Z‐score of the inflammation panel for each time point.

Data information: Significant proteins are displayed by red dots, and non‐significant ones with gray dots. Red letters indicate inflammation factors that correlate with the BMI and that were used to generate panel (G). A Benjamini–Hochberg FDR of 0.05 was used for significance in all correlation analyses (A–D).
Figure 6. Insulin resistance and systemic inflammation
Figure 6. Insulin resistance and systemic inflammation
  1. The five proteins with the highest positive and the four proteins with the highest negative correlation with IR were used to define a pro‐ and an anti‐IR panel. These panels separated the study cohort in a high and a low IR group and the 24 individuals that were present in at least one of the IR panels are highlighted (numbered in red at the y‐axes). Likewise, the ten‐protein inflammation panel separates the cohort in individuals with high and low systemic inflammation levels and individuals with high levels were highlighted (numbered in red). Of 16 study participants with high inflammation levels, 14 were also present in the high IR group as illustrated by a Venn diagram, indicating a high metabolic burden group as defined by plasma proteome profiling.

  2. HOMA‐IR levels and the BMI are compared between the high and the low metabolic burden group with indicated changes in percent at the study endpoint. These changes are linked to longitudinal changes in the anti‐IR, the pro‐IR, and the inflammation profile. The means are plotted with SEM as error bars over time.

Figure 7. Effect of weight loss on…
Figure 7. Effect of weight loss on the apolipoprotein family
  1. The initial LFQ intensity before weight loss (week −8) was set to 100% to normalize protein abundance within each participant to account for individual‐specific protein levels. Colors derive from clusters in Fig 4. The means are plotted with SEM as error bars over time. For the acute phase protein SAA1, the peak at week 13 was caused by very high levels in one individual (gray curve and right y‐axis in the 6th panel). Excluding this individual results in the red curve. Numbers below the protein name refer to their presence in the lipoprotein particle in panel (C).

  2. Annotation for gene ontology cellular component (GOCC) was Z‐scored and filtered for main lipoprotein particles.

  3. Gene ontology biological process (GOBP) annotations were filtered for the keywords lipid, lipoprotein, fat, and cholesterol, leading to the displayed group of GOBPs, which decreased due to weight loss.

Figure EV3. MS ‐measured protein intensities compared…
Figure EV3. MS‐measured protein intensities compared to standard clinical measurements
  1. Correlation of APOA1 with HDL (according to the literature each HDL particle should contain approximately one APOA1 molecule). The imperfect correlation can partially be explained by the lack of resolution of the reported HDL values, which can be seen by the non‐continuous values for the individuals, resulting in vertical lines.

  2. MS‐measured APOB intensities were correlated with LDL values (each LDL particle should contain one APOB molecule).

  3. Correlation of APOB intensities with cholesterol measurements.

Figure EV4. Hierarchical clustering of lipid metabolism‐related…
Figure EV4. Hierarchical clustering of lipid metabolism‐related GOBP terms
GOBP terms annotations were Z‐scored over time and filtered for the keywords lipid, lipoprotein, fat, and cholesterol. Hierarchical clustering indicates lipid metabolism‐related biological processes in response to weight loss and maintenance.

References

    1. Aebersold R, Mann M (2016) Mass‐spectrometric exploration of proteome structure and function. Nature 537: 347–355
    1. Anderson NL (2010) The clinical plasma proteome: a survey of clinical assays for proteins in plasma and serum. Clin Chem 56: 177–185
    1. Anderson NL (2014) Six decades searching for meaning in the proteome. J Proteomics 107: 24–30
    1. Azrad M, Gower BA, Hunter GR, Nagy TR (2012) Intra‐abdominal adipose tissue is independently associated with sex‐hormone binding globulin in premenopausal women. Obesity (Silver Spring) 20: 1012–1015
    1. Bozaoglu K, Attard C, Kulkarni H, Cummings N, Diego VP, Carless MA, Shields KA, Johnson MP, Kowlessur S, Dyer TD, Comuzzie AG, Almasy L, Zimmet P, Moses EK, Goring HH, Curran JE, Blangero J, Jowett JB (2014) Plasma levels of soluble interleukin 1 receptor accessory protein are reduced in obesity. J Clin Endocrinol Metab 99: 3435–3443
    1. Carlsson L, Lind L, Larsson A (2010) Reference values for 27 clinical chemistry tests in 70‐year‐old males and females. Gerontology 56: 259–265
    1. Casazza K, Pate R, Allison DB (2013) Myths, presumptions, and facts about obesity. N Engl J Med 368: 2236–2237
    1. Ceron JJ, Tecles F, Tvarijonaviciute A (2014) Serum paraoxonase 1 (PON1) measurement: an update. BMC Vet Res 10: 74
    1. Cominetti O, Nunez Galindo A, Corthesy J, Oller Moreno S, Irincheeva I, Valsesia A, Astrup A, Saris WH, Hager J, Kussmann M, Dayon L (2016) Proteomic biomarker discovery in 1000 human plasma samples with mass spectrometry. J Proteome Res 15: 389–399
    1. Cox J, Mann M (2008) MaxQuant enables high peptide identification rates, individualized p.p.b.‐range mass accuracies and proteome‐wide protein quantification. Nat Biotechnol 26: 1367–1372
    1. Cox J, Neuhauser N, Michalski A, Scheltema RA, Olsen JV, Mann M (2011) Andromeda: a peptide search engine integrated into the MaxQuant environment. J Proteome Res 10: 1794–1805
    1. Cox J, Hein MY, Luber CA, Paron I, Nagaraj N, Mann M (2014) Accurate proteome‐wide label‐free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol Cell Proteomics 13: 2513–2526
    1. Crawford PT, Elisens WJ (2006) Genetic variation and reproductive system among North American species of Nuttallanthus (Plantaginaceae). Am J Bot 93: 582–591
    1. Dominiczak MH, Caslake MJ (2011) Apolipoproteins: metabolic role and clinical biochemistry applications. Ann Clin Biochem 48: 498–515
    1. Duke‐Cohan JS, Gu J, McLaughlin DF, Xu Y, Freeman GJ, Schlossman SF (1998) Attractin (DPPT‐L), a member of the CUB family of cell adhesion and guidance proteins, is secreted by activated human T lymphocytes and modulates immune cell interactions. Proc Natl Acad Sci USA 95: 11336–11341
    1. Eckel RH, Grundy SM, Zimmet PZ (2005) The metabolic syndrome. Lancet 365: 1415–1428
    1. Esser N, Legrand‐Poels S, Piette J, Scheen AJ, Paquot N (2014) Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract 105: 141–150
    1. Famulla S, Lamers D, Hartwig S, Passlack W, Horrighs A, Cramer A, Lehr S, Sell H, Eckel J (2011) Pigment epithelium‐derived factor (PEDF) is one of the most abundant proteins secreted by human adipocytes and induces insulin resistance and inflammatory signaling in muscle and fat cells. Int J Obes (Lond) 35: 762–772
    1. Geyer PE, Kulak NA, Pichler G, Holdt LM, Teupser D, Mann M (2016) Plasma proteome profiling to assess human health and disease. Cell Syst 2: 185–195
    1. Goodpaster BH, Delany JP, Otto AD, Kuller L, Vockley J, South‐Paul JE, Thomas SB, Brown J, McTigue K, Hames KC, Lang W, Jakicic JM (2010) Effects of diet and physical activity interventions on weight loss and cardiometabolic risk factors in severely obese adults: a randomized trial. JAMA 304: 1795–1802
    1. van Greevenbroek MM, Schalkwijk CG, Stehouwer CD (2013) Obesity‐associated low‐grade inflammation in type 2 diabetes mellitus: causes and consequences. Neth J Med 71: 174–187
    1. Grundy SM (2015) Adipose tissue and metabolic syndrome: too much, too little or neither. Eur J Clin Invest 45: 1209–1217
    1. Hansen BA, Bray GA (2008) The metabolic syndrome: epidemiology, clinical treatment, and underlying mechanisms. Totowa, NJ: Humana Press;
    1. Hirai K, Hussey HJ, Barber MD, Price SA, Tisdale MJ (1998) Biological evaluation of a lipid‐mobilizing factor isolated from the urine of cancer patients. Cancer Res 58: 2359–2365
    1. Iepsen EW, Lundgren J, Dirksen C, Jensen JE, Pedersen O, Hansen T, Madsbad S, Holst JJ, Torekov SS (2015) Treatment with a GLP‐1 receptor agonist diminishes the decrease in free plasma leptin during maintenance of weight loss. Int J Obes (Lond) 39: 834–841
    1. Kamstrup PR, Benn M, Tybjaerg‐Hansen A, Nordestgaard BG (2008) Extreme lipoprotein(a) levels and risk of myocardial infarction in the general population: the Copenhagen City Heart Study. Circulation 117: 176–184
    1. Kulak NA, Pichler G, Paron I, Nagaraj N, Mann M (2014) Minimal, encapsulated proteomic‐sample processing applied to copy‐number estimation in eukaryotic cells. Nat Methods 11: 319–324
    1. Kushner RF, Ryan DH (2014) Assessment and lifestyle management of patients with obesity: clinical recommendations from systematic reviews. JAMA 312: 943–952
    1. Laudes M, Oberhauser F, Schulte DM, Schilbach K, Freude S, Bilkovski R, Schulz O, Faust M, Krone W (2010) Dipeptidyl‐peptidase 4 and attractin expression is increased in circulating blood monocytes of obese human subjects. Exp Clin Endocrinol Diabetes 118: 473–477
    1. Laugerette F, Alligier M, Bastard JP, Drai J, Chanseaume E, Lambert‐Porcheron S, Laville M, Morio B, Vidal H, Michalski MC (2014) Overfeeding increases postprandial endotoxemia in men: inflammatory outcome may depend on LPS transporters LBP and sCD14. Mol Nutr Food Res 58: 1513–1518
    1. Lewis JG, Bagley CJ, Elder PA, Bachmann AW, Torpy DJ (2005) Plasma free cortisol fraction reflects levels of functioning corticosteroid‐binding globulin. Clin Chim Acta 359: 189–194
    1. Li XA, Yutani C, Shimokado K (1998) Serum amyloid P component associates with high density lipoprotein as well as very low density lipoprotein but not with low density lipoprotein. Biochem Biophys Res Commun 244: 249–252
    1. Liu Y, Buil A, Collins BC, Gillet LC, Blum LC, Cheng LY, Vitek O, Mouritsen J, Lachance G, Spector TD, Dermitzakis ET, Aebersold R (2015) Quantitative variability of 342 plasma proteins in a human twin population. Mol Syst Biol 11: 786
    1. Look ARG, Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, Espeland MA, Evans M, Foreyt JP, Ghazarian S, Gregg EW, Harrison B, Hazuda HP, Hill JO, Horton ES, Hubbard VS et al (2013) Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med 369: 145–154
    1. Malmstrom E, Kilsgard O, Hauri S, Smeds E, Herwald H, Malmstrom L, Malmstrom J (2016) Large‐scale inference of protein tissue origin in gram‐positive sepsis plasma using quantitative targeted proteomics. Nat Commun 7: 10261
    1. Mann M, Kulak NA, Nagaraj N, Cox J (2013) The coming age of complete, accurate, and ubiquitous proteomes. Mol Cell 49: 583–590
    1. McEneny J, Daniels JA, McGowan A, Gunness A, Moore K, Stevenson M, Young IS, Gibney J (2015) A cross‐sectional study demonstrating increased serum amyloid a related inflammation in high‐density lipoproteins from subjects with type 1 diabetes mellitus and how this association was augmented by poor Glycaemic control. J Diabetes Res 2015: 351601
    1. Munoz J, Heck AJ (2014) From the human genome to the human proteome. Angew Chem 53: 10864–10866
    1. Nagaraj N, Kulak NA, Cox J, Neuhauser N, Mayr K, Hoerning O, Vorm O, Mann M (2012) System‐wide perturbation analysis with nearly complete coverage of the yeast proteome by single‐shot ultra HPLC runs on a bench top Orbitrap. Mol Cell Proteomics 11: M111.013722
    1. Newgard CB (2016) Metabolomics and metabolic diseases: where do we stand? Cell Metab doi:
    1. Pellet‐Many C, Frankel P, Jia H, Zachary I (2008) Neuropilins: structure, function and role in disease. Biochem J 411: 211–226
    1. Piccolo BD, Keim NL, Fiehn O, Adams SH, Van Loan MD, Newman JW (2015) Habitual physical activity and plasma metabolomic patterns distinguish individuals with low vs. high weight loss during controlled energy restriction. J Nutr 145: 681–690
    1. Ridker PM, Rifai N, Rose L, Buring JE, Cook NR (2002) Comparison of C‐reactive protein and low‐density lipoprotein cholesterol levels in the prediction of first cardiovascular events. N Engl J Med 347: 1557–1565
    1. Riecke BF, Christensen R, Christensen P, Leeds AR, Boesen M, Lohmander LS, Astrup A, Bliddal H (2010) Comparing two low‐energy diets for the treatment of knee osteoarthritis symptoms in obese patients: a pragmatic randomized clinical trial. Osteoarthritis Cartilage 18: 746–754
    1. Scheltema RA, Mann M (2012) SprayQc: a real‐time LC‐MS/MS quality monitoring system to maximize uptime using off the shelf components. J Proteome Res 11: 3458–3466
    1. Scheltema RA, Hauschild JP, Lange O, Hornburg D, Denisov E, Damoc E, Kuehn A, Makarov A, Mann M (2014) The Q Exactive HF, a Benchtop mass spectrometer with a pre‐filter, high‐performance quadrupole and an ultra‐high‐field Orbitrap analyzer. Mol Cell Proteomics 13: 3698–3708
    1. Selvin E, Paynter NP, Erlinger TP (2007) The effect of weight loss on C‐reactive protein: a systematic review. Arch Intern Med 167: 31–39
    1. Seres I, Bajnok L, Harangi M, Sztanek F, Koncsos P, Paragh G (2010) Alteration of PON1 activity in adult and childhood obesity and its relation to adipokine levels. Adv Exp Med Biol 660: 129–142
    1. Smith DE, Hanna R, Della F, Moore H, Chen H, Farese AM, MacVittie TJ, Virca GD, Sims JE (2003) The soluble form of IL‐1 receptor accessory protein enhances the ability of soluble type II IL‐1 receptor to inhibit IL‐1 action. Immunity 18: 87–96
    1. Soker S, Takashima S, Miao HQ, Neufeld G, Klagsbrun M (1998) Neuropilin‐1 is expressed by endothelial and tumor cells as an isoform‐specific receptor for vascular endothelial growth factor. Cell 92: 735–745
    1. Surinova S, Schiess R, Huttenhain R, Cerciello F, Wollscheid B, Aebersold R (2011) On the development of plasma protein biomarkers. J Proteome Res 10: 5–16
    1. Thompson PA, Tobias PS, Viriyakosol S, Kirkland TN, Kitchens RL (2003) Lipopolysaccharide (LPS)‐binding protein inhibits responses to cell‐bound LPS. J Biol Chem 278: 28367–28371
    1. Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein M, Geiger T, Mann M, Cox J (2016) The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods 13: 731–740
    1. Utermann G (1989) The mysteries of lipoprotein(a). Science 246: 904–910
    1. Wahl S, Vogt S, Stuckler F, Krumsiek J, Bartel J, Kacprowski T, Schramm K, Carstensen M, Rathmann W, Roden M, Jourdan C, Kangas AJ, Soininen P, Ala‐Korpela M, Nothlings U, Boeing H, Theis FJ, Meisinger C, Waldenberger M, Suhre K et al (2015) Multi‐omic signature of body weight change: results from a population‐based cohort study. BMC Med 13: 48
    1. Wang P, Mariman E, Keijer J, Bouwman F, Noben JP, Robben J, Renes J (2004) Profiling of the secreted proteins during 3T3‐L1 adipocyte differentiation leads to the identification of novel adipokines. Cell Mol Life Sci 61: 2405–2417
    1. Wang P, Smit E, Brouwers MC, Goossens GH, van der Kallen CJ, van Greevenbroek MM, Mariman EC (2008) Plasma pigment epithelium‐derived factor is positively associated with obesity in Caucasian subjects, in particular with the visceral fat depot. Eur J Endocrinol 159: 713–718
    1. Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE, Tataranni PA (2001) Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab 86: 1930–1935
    1. Yudkin JS, Stehouwer CD, Emeis JJ, Coppack SW (1999) C‐reactive protein in healthy subjects: associations with obesity, insulin resistance, and endothelial dysfunction: a potential role for cytokines originating from adipose tissue? Arterioscler Thromb Vasc Biol 19: 972–978
    1. Zubarev RA, Makarov A (2013) Orbitrap mass spectrometry. Anal Chem 85: 5288–5296

Source: PubMed

3
S'abonner