F. Valeria-d'argenio and . Salvatore, The role of the gut microbiome in the healthy adult status". en, Clinica Chimica Acta, vol.451, 2015.

, Structure, function and diversity of the healthy human microbiome". en, The Human Microbiome Project Consortium, vol.486, pp.28-0836, 2012.

M. Arumugam, Enterotypes of the human gut microbiome, Nature, vol.473, pp.28-0836, 2011.
URL : https://hal.archives-ouvertes.fr/cea-00903625

L. Chatelier-emmanuelle, Richness of human gut microbiome correlates with metabolic markers". en, Nature, vol.500, pp.28-0836, 2013.

J. Li, An integrated catalog of reference genes in the human gut microbiome". en, Nature Biotechnology, vol.32, pp.1087-0156, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01195478

N. Sven-bastiaan-haange and . Jehmlich, Proteomic interrogation of the gut microbiota: potential clinical impact". en, Expert Review of Proteomics, vol.13, issue.6, pp.1478-9450, 2016.

Y. Pey and . Lee, Metaproteomic analysis of human gut microbiota: where are we heading?" en, Journal of Biomedical Science, vol.24, issue.1, p.36, 2017.

W. Xiong, Microbial metaproteomics for characterizing the range of metabolic functions and activities of human gut microbiota". en, pp.3424-3438, 2015.

X. Zhang and D. Figeys, Perspective and Guidelines for Metaproteomics in Microbiome Studies". en, Journal of Proteome Research, vol.18, issue.6, pp.1535-3893, 2019.

S. Deusch, News in livestock research -use of Omics -technologies to study the microbiota in the gastrointestinal tract of farm animals". en, Computational and Structural Biotechnology Journal, vol.13, pp.55-63, 2015.

P. Gerren and . Hobby, Chronic kidney disease and the gut microbiome". en, American Journal of Physiology-Renal Physiology, vol.316, issue.6, 2018.

R. I. Amann, W. Ludwig, and K. H. Schleifer, Phylogenetic identification and in situ detection of individual microbial cells without cultivation". eng, Microbiological Reviews, vol.59, issue.1, pp.146-0749, 1995.

P. Hugenholtz, M. Brett, N. Goebel, and . Pace, Impact of Culture-Independent Studies on the Emerging Phylogenetic View of Bacterial Diversity". en, J. BACTERIOL, vol.180, p.10, 1998.

E. Allen-vercoe, Bringing the gut microbiota into focus through microbial culture: recent progress and future perspective". en, Current Opinion in Microbiology, vol.16, issue.5, pp.625-629, 2013.

J. Bodilis, Variable Copy Number, Intra-Genomic Heterogeneities and Lateral Transfers of the 16S rRNA Gene in Pseudomonas". en, PLoS ONE, vol.7, issue.4, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00736976

J. Jovel, Characterization of the Gut Microbiome Using 16S or Shotgun Metagenomics, Frontiers in Microbiology, vol.7, 2016.

L. Chistoserdova, Functional Metagenomics: Recent Advances and Future Challenges". en, vol.26, pp.264-8725, 2009.

, Principe de la source ionisation électrospray, 2016.

T. Fisher, Orbitrap :: Orbitrap Fusion Lumos

D. N. Perkins, Probability-based protein identification by searching sequence databases using mass spectrometry data". en, Electrophoresis, vol.20, pp.3551-3567, 1999.

,

Y. Lewis and . Geer, Open Mass Spectrometry Search Algorithm". en. In: Journal of Proteome Research, vol.3, issue.5, pp.1535-3893, 2004.

R. Craig and . Beavis, TANDEM: matching proteins with tandem mass spectra, In: Bioinformatics, vol.20, pp.1367-4803, 2004.

C. Blank, Disseminating Metaproteomic Informatics Capabilities and Knowledge Using the Galaxy-P Framework". en, Proteomes 6.1, pp.2227-7382, 2018.

M. Brosch, Comparison of Mascot and X!Tandem Performance for Low and High Accuracy Mass Spectrometry and the Development of an Adjusted Mascot Threshold". en, Molecular & Cellular Proteomics, vol.7, issue.5, pp.1535-9476, 2008.

D. Zosso, Tandem mass spectrometry protein identification on a PC grid, Studies in Health Technology and Informatics, vol.126, pp.926-9630, 2007.

M. N. Akhtar, Evaluation of Database Search Programs for Accurate Detection of Neuropeptides in Tandem Mass Spectrometry Experiments". en, Journal of Proteome Research, vol.11, pp.1535-3893, 2012.

S. Aggarwal and A. Kumar-yadav, False Discovery Rate Estimation in Proteomics, Statistical Analysis in Proteomics

, , vol.1362, pp.119-128, 2016.

L. Käll, Assigning Significance to Peptides Identified by Tandem Mass Spectrometry Using Decoy Databases". en, Journal of Proteome Research, vol.7, pp.1535-3893, 2008.

O. Langella, X!TandemPipeline: A Tool to Manage Sequence Redundancy for Protein Inference and Phosphosite Identification". en, Journal of Proteome Research, vol.16, pp.1535-3893, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01484169

C. Juste, Bacterial protein signals are associated with Crohn's disease". en, Gut 63, vol.10, pp.17-5749, 2014.

M. Blein-nicolas, Including shared peptides for estimating protein abundances: A significant improvement for quantitative proteomics". en, PROTEOMICS 12, vol.18, pp.2797-2801, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02645398

M. Blein, -. , and M. Zivy, Thousand and one ways to quantify and compare protein abundances in label-free bottom-up proteomics". en, Biochimica et Biophysica Acta (BBA) -Proteins and Proteomics, vol.1864, pp.883-895, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01532597

P. Wilmes and P. L. Bond, The application of two-dimensional polyacrylamide gel electrophoresis and downstream analyses to a mixed community of prokaryotic microorganisms". en, Environmental Microbiology, vol.6, issue.9, pp.1462-2912, 2004.

. Rj-ram, Community proteomics of a natural microbial biofilm, In: Science, vol.308, 2005.

E. S. Klaassens, W. M. De-vos, and E. E. Vaughan, Metaproteomics Approach To Study the Functionality of the Microbiota in the Human Infant Gastrointestinal, Tract". en. In: Applied and Environmental Microbiology, vol.73, pp.99-2240, 2007.

C. A. Kolmeder, Comparative Metaproteomics and Diversity Analysis of Human Intestinal Microbiota Testifies for Its Temporal Stability and Expression of Core Functions". en, PLoS ONE, vol.7, issue.1, 2012.

K. Rooijers, An iterative workflow for mining the human intestinal metaproteome". en, BMC Genomics, vol.12, issue.1, pp.1471-2164, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00656193

E. Hernández, Functional consequences of microbial shifts in the human gastrointestinal tract linked to antibiotic treatment and obesity". en, Gut Microbes, vol.4, pp.1949-0976, 2013.

A. Tanca, Potential and active functions in the gut microbiota of a healthy human cohort". en, Microbiome 5.1, pp.2049-2618, 2017.

A. Tanca, The impact of sequence database choice on metaproteomic results in gut microbiota studies, pp.2049-2618, 2016.

X. Zhang, MetaPro-IQ: a universal metaproteomic approach to studying human and mouse gut microbiota, 2016.

, , pp.2049-2618

A. Tanca, Enrichment or depletion? The impact of stool pretreatment on metaproteomic characterization of the human gut microbiota". en, PROTEOMICS 15, vol.20, 2015.

C. S. Reigstad and P. C. Kashyap, Beyond phylotyping: understanding the impact of gut microbiota on host biology, en. In: Neurogastroenterology & Motility, vol.25, pp.358-372, 2013.

A. Heintz-buschart, Integrated multi-omics of the human gut microbiome in a case study of familial type 1 diabetes". en, Nature Microbiology, vol.2, issue.1, pp.2058-5276, 2017.

R. D. Zwittink, Metaproteomics reveals functional differences in intestinal microbiota development of preterm infants". en, Molecular & Cellular Proteomics, vol.16, pp.1535-9476, 2017.

B. Brooks, Strain-resolved microbial community proteomics reveals simultaneous aerobic and anaerobic function during gastrointestinal tract colonization of a preterm infant, Frontiers in Microbiology, vol.6, 2015.

M. Guirro, Multi-omics approach to elucidate the gut microbiota activity: Metaproteomics and metagenomics connection". en, pp.1692-1701, 2018.

W. Xiong, Genome-resolved metaproteomic characterization of preterm infant gut microbiota development reveals species-specific metabolic shifts and variabilities during early life". en, Microbiome 5.1, pp.2049-2618, 2017.

J. C. Young, Metaproteomics reveals functional shifts in microbial and human proteins during a preterm infant gut colonization case". en, PROTEOMICS 15, vol.20, pp.3463-3473, 2015.

, , 2019.

S. Haange, Metaproteome Analysis and Molecular Genetics of Rat Intestinal Microbiota Reveals Section and Localization Resolved Species Distribution and Enzymatic Functionalities". en, Journal of Proteome Research, vol.11, issue.11, pp.1535-3893, 2012.

S. Levi-mortera, Metaproteomic investigation to assess gut microbiota shaping in newborn mice: A combined taxonomic, functional and quantitative approach". en, Journal of Proteomics, vol.203, 2019.

X. Ke, Synbiotic-driven improvement of metabolic disturbances is associated with changes in the gut microbiome in diet-induced obese mice". en, Molecular Metabolism, vol.22, pp.96-109, 2019.

M. Schaubeck, Dysbiotic gut microbiota causes transmissible Crohn's disease-like ileitis independent of failure in antimicrobial defence". en, Gut 65, pp.17-5749, 2016.

A. Tanca, Caloric restriction promotes functional changes involving short-chain fatty acid biosynthesis in the rat gut microbiota". en, Scientific Reports, vol.8, issue.1, pp.2045-2322, 2018.

J. Tröscher-mußotter, Analysis of the Bacterial and Host Proteins along and across the Porcine Gastrointestinal Tract". en, Proteomes 7.1, p.4, 2019.

D. Borda-molina, J. Seifert, and A. Camarinha-silva, Current Perspectives of the Chicken Gastrointestinal Tract and Its Microbiome". en, Computational and Structural Biotechnology Journal, vol.16, pp.131-139, 2018.

A. Palomba, Multi-Omic Biogeography of the Gastrointestinal Microbiota of a Pre-Weaned Lamb". en, Proteomes 5, vol.4, p.36, 2017.

B. Tilocca, Variations of Phosphorous Accessibility Causing Changes in Microbiome Functions in the Gastrointestinal Tract of Chickens". en, PLOS ONE, vol.11, issue.10, 2016.

Y. Tang, Metaproteomics Analysis Reveals the Adaptation Process for the Chicken Gut Microbiota". en, Applied and Environmental Microbiology, vol.80, issue.2, pp.99-2240, 2014.

R. H. Mills, Evaluating Metagenomic Prediction of the Metaproteome in a 4.5-Year Study of a Patient with Crohn's Disease". en, pp.2379-5077, 2019.

X. Zhang, Metaproteomics reveals associations between microbiome and intestinal extracellular vesicle proteins in pediatric inflammatory bowel disease". en, Nature Communications, vol.9, issue.1, pp.2041-1723, 2018.

A. D. Kostic, R. J. Xavier, and D. Gevers, The Microbiome in Inflammatory Bowel Disease: Current Status and the Future Ahead". en, Gastroenterology, vol.146, issue.6, pp.1489-1499, 2014.

A. Vich-vila, Gut microbiota composition and functional changes in inflammatory bowel disease and irritable bowel syndrome, en. In: Science Translational Medicine, vol.10, 2018.

X. Zhang, Assessing the impact of protein extraction methods for human gut metaproteomics". en, Journal of Proteomics, vol.180, pp.120-127, 2018.

T. Muth, Navigating through metaproteomics data: A logbook of database searching". en, PROTEOMICS 15, vol.20, pp.3439-3453, 2015.

, , 2018.

M. Shao-en-ong and . Mann, Mass spectrometry-based proteomics turns quantitative". en, Nature Chemical Biology, vol.1, issue.5, pp.1552-4450, 2005.

C. Chang, LFAQ: towards unbiased label-free absolute protein quantification by predicting peptide quantitative factors, Bioinformatics, 2018.

B. He, Label-free absolute protein quantification with data-independent acquisition". en, Journal of Proteomics, vol.200, pp.51-59, 2019.

C. Nathan and . Verberkmoes, Shotgun metaproteomics of the human distal gut microbiota". en, The ISME Journal, vol.3, pp.1751-7362, 2009.

E. J. Benjamin, Update: A Report From the American Heart Association". en. In: Circulation, vol.135, 2017.

K. George, M. M. Alberti, P. Zimmet, and J. Shaw, The metabolic syndrome-a new worldwide definition". en, The Lancet, vol.366, pp.1059-1062, 2005.

M. Glorian and I. Limon, L'athérosclérose, une maladie inflammatoire". fr. In: Revue Francophone des Laboratoires, vol.389, pp.43-48, 2007.

J. Fruchart, New Risk Factors for Atherosclerosis and Patient Risk Assessment". en, Circulation 109.23_suppl_1, 2004.

A. P. Goldberg, Aerobic and resistive exercise modify risk factors for coronary heart disease:" en, Medicine & Science in Sports & Exercise, vol.21, issue.6, pp.195-9131, 1989.

N. P. Larsen, Diet and Atherosclerosis: A Field Study". en, A.M.A. Archives of Internal Medicine, vol.100, issue.3, pp.888-2479, 1957.

A. Keys, Human Atherosclerosis and the Diet". en, Circulation 5.1, pp.9-7322, 1952.

. Marco-matteo-ciccone, Dietary Intake of Carotenoids and Their Antioxidant and Anti-Inflammatory Effects in Cardiovascular Care". en, Mediators of Inflammation 2013, pp.962-9351, 2013.

V. Summerhill, Vasculoprotective Role of Olive Oil Compounds via Modulation of Oxidative Stress in Atherosclerosis, In: Frontiers in Cardiovascular Medicine, vol.5, 2018.

C. Mercado and E. A. Jaimes, Cigarette smoking as a risk factor for atherosclerosis and renal disease: Novel pathogenic insights". en, Current Hypertension Reports, vol.9, issue.1, pp.1522-6417, 2007.

T. H. Dent, Predicting the risk of coronary heart disease". en, Atherosclerosis, vol.213, pp.345-351, 2010.

H. Tilg and A. Kaser, Gut microbiome, obesity, and metabolic dysfunction". en, Journal of Clinical Investigation, vol.121, issue.6, pp.21-9738, 2011.

N. Larsen, Gut Microbiota in Human Adults with Type 2 Diabetes Differs from Non-Diabetic Adults". en, PLoS ONE, vol.5, 2010.

, , 2017.

B. Sartor and . Sarkis-k-mazmanian, Intestinal Microbes in Inflammatory Bowel Diseases, The American Journal of Gastroenterology Supplements, vol.1, issue.1, pp.1948-9498, 2012.

R. E. Ley, Human gut microbes associated with obesity". en, Nature, vol.444, pp.28-0836, 2006.

J. Qin, A metagenome-wide association study of gut microbiota in type 2 diabetes, Nature, vol.490, pp.28-0836, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01204262

A. Cotillard, Dietary intervention impact on gut microbial gene richness, Nature, vol.500, pp.28-0836, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01001543

. Ling-chun-kong, Insulin resistance and inflammation predict kinetic body weight changes in response to dietary weight loss and maintenance in overweight and obese subjects by using a Bayesian network approach". en, The American Journal of Clinical Nutrition, vol.98, issue.6, pp.1385-1394, 2013.

, , pp.2-9165

P. Gerard, Gnotobiotic rats harboring human intestinal microbiota as a model for studying cholesterol-to-coprostanol conversion". en, FEMS Microbiology Ecology, vol.47, issue.3, p.1686496, 2004.
URL : https://hal.archives-ouvertes.fr/hal-01606922

P. Gerard, Bacteroides sp. Strain D8, the First Cholesterol-Reducing Bacterium Isolated from, Human Feces". en. In: Applied and Environmental Microbiology, vol.73, pp.5742-5749, 2007.
URL : https://hal.archives-ouvertes.fr/hal-01608615

F. Bäckhed, Mechanisms underlying the resistance to diet-induced obesity in germ-free mice". en, Proceedings of the National Academy of Sciences, vol.104, issue.3, pp.27-8424, 2007.

, Gut organisms could be clue in controlling obesity risk, In: ScienceDaily, 2012.

N. Alang and C. R. Kelly, Weight Gain After Fecal Microbiota Transplantation". en, Open Forum Infectious Diseases, vol.2, pp.4-004, 2015.

, , pp.2328-8957

F. Backhed, The gut microbiota as an environmental factor that regulates fat storage". en, Proceedings of the National Academy of Sciences, vol.101, pp.27-8424, 2004.

J. Peter and . Turnbaugh, An obesity-associated gut microbiome with increased capacity for energy harvest". en, Nature, vol.444, pp.28-0836, 2006.

Z. Wang, Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease, Nature, vol.472, pp.28-0836, 2011.

H. Fredrik and . Karlsson, Symptomatic atherosclerosis is associated with an altered gut metagenome". en, Nature Communications, vol.3, issue.1, 2012.

, , pp.2041-1723

J. Rechenberger, Challenges in Clinical Metaproteomics Highlighted by the Analysis of Acute Leukemia Patients with Gut Colonization by Multidrug-Resistant Enterobacteriaceae". en, Proteomes 7.1, pp.2227-7382, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02262589

J. Qin, A human gut microbial gene catalogue established by metagenomic sequencing, Nature, vol.464, pp.28-0836, 2010.
URL : https://hal.archives-ouvertes.fr/cea-00908974

P. Jagtap, Deep metaproteomic analysis of human salivary supernatant". en, PROTEOMICS 12, vol.7, pp.992-1001, 2012.

P. Jagtap, A two-step database search method improves sensitivity in peptide sequence matches for metaproteomics and proteogenomics studies". en, PROTEOMICS 13, vol.8, pp.1352-1357, 2013.

K. Cheng, MetaLab: an automated pipeline for metaproteomic data analysis, Microbiome 5, pp.2049-2618, 2017.

D. Beyter, ProteoStorm: An Ultrafast Metaproteomics Database Search Framework". en, Cell Systems, vol.7, pp.463-467, 2018.

T. C. Hsieh, K. H. Ma, and A. Chao, iNEXT: an R package for rarefaction and extrapolation of species diversity (Hill numbers)". en, Methods in Ecology and Evolution 7.12 (Dec. 2016). Ed. by Greg McInerny, pp.1451-1456

. R-core-team, R: A Language and Environment for Statistical Computing, 2013.

D. L. Tabb, Repeatability and Reproducibility in Proteomic Identifications by Liquid Chromatography-Tandem Mass Spectrometry". en, Journal of Proteome Research, vol.9, issue.2, pp.1535-3893, 2010.

N. M. Delzenne and P. D. Cani, Interaction Between Obesity and the Gut Microbiota: Relevance in Nutrition". en, Annual Review of Nutrition, vol.31, issue.1, pp.199-9885, 2011.

D. Brian, . Ondov, H. Nicholas, A. M. Bergman, and . Phillippy, Interactive metagenomic visualization in a Web browser". en, BMC Bioinformatics, vol.12, issue.1, pp.1471-2105, 2011.

M. Fava, Weight gain and antidepressants". eng, The Journal of Clinical Psychiatry, vol.61, issue.11, pp.160-6689, 2000.

M. Lieke and . Spekhorst, Performance of the Montreal classification for inflammatory bowel diseases". en, In: World Journal of Gastroenterology, vol.20, pp.1007-9327, 2014.

B. Mattos, Inflammatory Bowel Disease: An Overview of Immune Mechanisms and Biological Treatments". en, Mediators of Inflammation 2015, pp.962-9351, 2015.

C. Daniel and . Baumgart, The Diagnosis and Treatment of Crohn's Disease and Ulcerative Colitis, Deutsches Aerzteblatt Online, 2009.

C. Abraham and J. H. Cho, Inflammatory Bowel Disease". en, New England Journal of Medicine, vol.361, pp.28-4793, 2009.

Q. He, Two distinct metacommunities characterize the gut microbiota in Crohn's disease patients". en, GigaScience 6, vol.7, pp.2047-217, 2017.

E. A. Franzosa, Gut microbiome structure and metabolic activity in inflammatory bowel disease". en, Nature Microbiology, vol.4, issue.2, pp.2058-5276, 2019.

A. Criscuolo and S. Brisse, AlienTrimmer: A tool to quickly and accurately trim off multiple short contaminant sequences from high-throughput sequencing reads". en, Genomics 102, pp.500-506, 2013.

B. Langmead, L. Steven, and . Salzberg, Fast gapped-read alignment with Bowtie 2". en, Nature Methods, vol.9, pp.1548-7091, 2012.

M. R. Crusoe, The khmer software package: enabling efficient nucleotide sequence analysis". en, pp.2046-1402, 2015.

A. Bankevich, SPAdes: A New Genome Assembly Algorithm and Its Applications to Single-Cell Sequencing". en, Journal of Computational Biology, vol.19, issue.5, pp.1066-5277, 2012.

S. Nurk, metaSPAdes: a new versatile metagenomic assembler". en, Genome Research, vol.27, pp.1088-9051, 2017.

E. Forouzan, Practical evaluation of 11 de novo assemblers in metagenome assembly". en, Journal of Microbiological Methods, vol.151, pp.99-105, 2018.

D. Hyatt, Prodigal: prokaryotic gene recognition and translation initiation site identification". en, BMC Bioinformatics, vol.11, issue.1, pp.1471-2105, 2010.

. William-l-trimble, Short-read reading-frame predictors are not created equal: sequence error causes loss of signal". en, BMC Bioinformatics, vol.13, issue.1, pp.1471-2105, 2012.

W. Li and A. Godzik, Cd-hit: a fast program for clustering and comparing large sets of protein or nucleotide sequences". en, Bioinformatics, vol.22, pp.1367-4803, 2006.

C. Wen, Quantitative metagenomics reveals unique gut microbiome biomarkers in ankylosing spondylitis". en, Genome Biology, vol.18, p.142, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02628835

B. L. Zybailov, Metaproteomics reveals potential mechanisms by which dietary resistant starch supplementation attenuates chronic kidney disease progression in rats". en, PLOS ONE, vol.14, 2019.

T. Chen, The Human Oral Microbiome Database: a web accessible resource for investigating oral microbe taxonomic and genomic information". en, Database 2010.0, 2010.

T. Yamada, iPath2.0: interactive pathway explorer, Nucleic Acids Res, vol.39, 2011.

J. and A. Blakeley-ruiz, Metaproteomics reveals persistent and phylumredundant metabolic functional stability in adult human gut microbiomes of Crohn's remission patients despite temporal variations in microbial taxa, genomes, and proteomes". en, Microbiome 7.1 (Dec. 2019), pp.2049-2618

C. Manichanh, The gut microbiota in IBD, Nature Reviews Gastroenterology & Hepatology, vol.9, pp.1759-5045, 2012.

X. Wang, MS1 ion current-based quantitative proteomics: A promising solution for reliable analysis of large biological cohorts". en, Mass Spectrometry Reviews, 2019.

B. Valot, MassChroQ: A versatile tool for mass spectrometry quantification". en, PROTEOMICS 11, vol.17, pp.1615-9861, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01481216

C. Lazar, Accounting for the Multiple Natures of Missing Values in Label-Free Quantitative Proteomics Data Sets to Compare Imputation Strategies". en, Journal of Proteome Research, vol.15, pp.1535-3893, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02083850

D. B. Rubin, Inference and missing data". en, Biometrika, vol.63, pp.6-3444, 1976.

T. Aittokallio, Dealing with missing values in large-scale studies: microarray data imputation and beyond". en, Briefings in Bioinformatics, vol.11, issue.2, pp.1467-5463, 2010.

A. Patel, Development and Evaluation of Statistical Approaches in Proteomic Biomarker Discovery, 2012.

J. M. Bobbie and . Webb-robertson, Review, Evaluation, and Discussion of the Challenges of Missing Value Imputation for Mass Spectrometry-Based Label-Free Global Proteomics". en, Journal of Proteome Research, vol.14, issue.5, pp.1535-3893, 2015.

P. Jonsson and C. Wohlin, An evaluation of k-nearest neighbour imputation using likert data, 10th International Symposium on Software Metrics, pp.108-118, 2004.

T. Metsalu and J. Vilo, ClustVis: a web tool for visualizing clustering of multivariate data using Principal Component Analysis and heatmap". en, W566-W570. ISSN: 0305-1048, vol.43, 2015.

Y. Yu, Comprehensive Metaproteomic Analyses of Urine in the Presence and Absence of Neutrophil-Associated Inflammation in the Urinary Tract". en, Theranostics 7, pp.238-252, 2017.

D. Mark, A. Robinson, and . Oshlack, A scaling normalization method for differential expression analysis of RNA-seq data". en, Genome Biology, vol.11, issue.3, 2010.

Y. Benjamini and Y. Hochberg, Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing". en, Journal of the Royal Statistical Society: Series B (Methodological), vol.57, issue.1, pp.289-300, 1995.

A. Reiner, D. Yekutieli, and Y. Benjamini, Identifying differentially expressed genes using false discovery rate controlling procedures". en, Bioinformatics, vol.19, issue.3, pp.1367-4803, 2003.

L. Breiman, Classification and regression based on a forest of trees using random inputs, Machine Learning, vol.45, pp.5-32, 2001.

M. Kuhn, Building Predictive Models in R Using the caret Package". en, Journal of Statistical Software, vol.28, 2008.

. #157-pcr, Biology Notes for A level, 2016.

E. Britannica, Transverse section of an artery, 2010.

A. Zmys?owski and A. Szterk, Current knowledge on the mechanism of atherosclerosis and pro-atherosclerotic properties of oxysterols". en, Lipids in Health and Disease, vol.16, issue.1, 2017.

P. Duriez, Mécanismes de formation de la plaque d'athérome". fr. In: La Revue de, Médecine Interne, vol.25, 2004.

A. C. Doran, N. Meller, and C. A. Mcnamara, Role of Smooth Muscle Cells in the Initiation and Early Progression of Atherosclerosis". en, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.28, issue.5, pp.1079-5642, 2008.

T. Seimon and I. Tabas, Mechanisms and consequences of macrophage apoptosis in atherosclerosis". en, Journal of Lipid Research, vol.50, pp.22-2275, 2009.

A. Tsipis, Novel Oral Anticoagulants in Peripheral Arterial and Coronary Artery Disease". en, Cardiovascular & Hematological Agents in Medicinal Chemistry, vol.12, issue.1, 2014.

P. Libby, M. Paul, G. K. Ridker, and . Hansson, Progress and challenges in translating the biology of atherosclerosis". en, Nature, vol.473, pp.28-0836, 2011.

J. Kultima, MOCAT: A Metagenomics Assembly and Gene Prediction Toolkit". en, PLoS ONE, vol.7, issue.10, 2012.

S. Highlander, Mock Community Analysis". en. In: Encyclopedia of Metagenomics, pp.1-7, 2014.

Y. Hou and S. Lin, Distinct Gene Number-Genome Size Relationships for Eukaryotes and Non-Eukaryotes: Gene Content Estimation for Dinoflagellate Genomes". en, PLoS ONE, vol.4, issue.9, 2009.