F. Bäckhed, R. E. Ley, J. L. Sonnenburg, D. A. Peterson, and J. I. Gordon, Host-bacterial mutualism in the human intestine, Science, vol.307, 1915.

S. S. Yoon, E. K. Kim, and W. J. Lee, Functional genomic and metagenomic approaches to understanding gut microbiota-animal mutualism, Curr. Opin. Microbiol, vol.24, pp.38-46, 2015.

R. L. Graham, C. Graham, and G. Mcmullan, Microbial proteomics: A mass spectrometry primer for biologists. Microb. Cell Fact, vol.6, pp.1-14, 2007.

P. Wilmes, P. L. Bond, and . Metaproteomics, Studying functional gene expression in microbial ecosystems, Trends Microbiol, vol.14, pp.92-97, 2006.

F. Rodríguez-valera, Environmental genomics, the big picture?, FEMS Microbiol. Lett, vol.231, pp.153-158, 2004.

J. S. Lichtman, A. Marcobal, J. L. Sonnenburg, and J. E. Elias, Host-centric proteomics of stool: A novel strategy focused on intestinal responses to the gut microbiota, Mol. Cell Proteom, vol.12, pp.3310-3318, 2013.

C. A. Kolmeder, M. De-been, J. Nikkilä, I. Ritamo, J. Mättö et al., Comparative metaproteomics and diversity analysis of human intestinal microbiota testifies for its temporal stability and expression of core functions, PLoS ONE, vol.7, 2012.

N. C. Verberkmoes, A. L. Russell, M. Shah, A. Godzik, M. Rosenquist et al., Shotgun metaproteomics of the human distal gut microbiota, vol.3, pp.179-189, 2009.

A. E. Pérez-cobas, M. J. Gosalbes, A. Friedrichs, H. Knecht, A. Artacho et al., Gut microbiota disturbance during antibiotic therapy: A multi-omic approach, Gut, vol.62, pp.1591-1601, 2012.

K. H. Chuong, D. R. Mack, A. Stintzi, and K. C. O'doeherty, Human Microbiome and Learning Healthcare Systems: Integrating Research and Precision Medicine for Inflammatory Bowel Disease, OMICS, vol.22, pp.119-126, 2018.

C. Juste, D. P. Kreil, C. Beauvallet, A. Guillot, S. Vaca et al., Bacterial protein signals are associated with Crohn's disease, Gut, vol.63, pp.1566-1577, 2014.

M. Ferrer, A. Riuz, F. Lanza, S. B. Haange, A. Oberbach et al., Microbiota from the distal guts of lean and obese adolescents exhibit partial functional redundancy besides clear differences in community structure, Environ. Microbiol, vol.15, pp.211-226, 2013.

C. A. Kolmeder, J. Ritari, F. J. Verdam, T. Muth, S. Keskitalo et al., Colonic metaproteomic signatures of active bacteria and the host in obesity, Proteomics, vol.15, pp.3544-3552, 2015.

P. G. Gavin, J. A. Mullaney, D. Loo, P. A. Gottlieb, M. M. Hill et al., Intestinal Metaproteomics Reveals Host-Microbiota Interactions in Subjects at Risk for Type 1 Diabetes, Diabetes Care, vol.41, pp.2178-2186, 2018.

S. P. Hammond and L. R. Baden, Antibiotic prophylaxis for patients with acute leukemia, Leuk Lymphoma, vol.49, pp.183-193, 2008.

R. Heyer, K. Schallert, R. Zoun, B. Becher, G. Saake et al., Challenges and perspectives of metaproteomic data analysis, J. Biotechnol, vol.261, pp.24-36, 2017.

E. Timmins-schiffman, D. H. May, M. Mikan, M. Riffle, C. Frazar et al., Critical decisions in metaproteomics: Achieving high confidence protein annotations in a sea of unknowns, vol.11, pp.309-314, 2017.

T. Muth, B. Y. Renard, and L. Martens, Metaproteomic data analysis at a glance: Advances in computational microbial community proteomics, Expert Rev. Proteom, vol.13, pp.757-769, 2016.

J. Lloyd-price, A. Mahurkar, G. Rahnavard, J. Crabtree, J. Orvis et al., Strains, functions and dynamics in the expanded Human Microbiome Project, Nature, vol.550, pp.61-66, 2017.

S. F. Altschul, W. Gish, W. Miller, E. W. Myers, and D. J. Lipman, Basic local alignment search tool, J. Mol. Biol, vol.215, pp.403-410, 1990.

M. Kleiner, E. Thorson, C. E. Sharp, X. Dong, D. Liu et al., Assessing species biomass contributions in microbial communities via metaproteomics, Nat. Commun, vol.8, 1558.

S. B. Haange and N. Jehmlich, Proteomic interrogation of the gut microbiota: Potential clinical impact, vol.13, pp.535-537, 2016.

A. P. Magiorakos, A. Srinivasan, R. B. Carey, Y. Carmeli, M. E. Falagas et al., Multidrug-resistant, extensively drug-resistant and pandrug-resistant bacteria: An international expert proposal for interim standard definitions for acquired resistance, Clin. Microbiol. Infect, vol.18, pp.268-281, 2014.

A. Shevchenko, H. Tomas, J. Halvis, B. Olsen, and M. Mann, In-gel digestion for mass spectrometric characterization of proteins and proteomes, Nat. Protoc, vol.1, pp.2856-2860, 2006.

H. Hahne, F. Pachl, B. Ruprecht, S. K. Maier, S. Klaeger et al., DMSO enhances electrospray response, boosting sensitivity of proteomic experiments, Nat. Methods, vol.10, pp.989-991, 2013.

J. Cox and M. Mann, MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification, Nat. Biotechnol, vol.26, pp.1367-1372, 2008.

J. Li, H. Jia, X. Cai, H. Zhong, Q. Feng et al., MetaHIT Consortium An integrated catalog of reference genes in the human gut microbiome, Nat. Biotechnol, vol.32, pp.834-841, 2014.

L. Käll, J. D. Canterbury, J. Weston, W. S. Noble, and M. J. Maccoss, Semi-supervised learning for peptide identification from shotgun proteomics datasets, Nat. Methods, vol.4, pp.923-925, 2007.

B. Mesuere, B. Devreese, G. Debyser, M. Aerts, P. Vandamme et al., Unipept: Tryptic peptide-based biodiversity analysis of metaproteome samples, J. Proteome Res, vol.11, pp.5773-5780, 2012.

S. R. Gurdeep, A. Tanca, A. Palomba, F. Van-der-jeugt, P. Verschaffelt et al., Unipept 4.0: Functional analysis of metaproteome data, J Proteome Res, 2018.

J. A. Vizcaíno, R. G. Côté, A. Csordas, J. A. Dianes, A. Fabregat et al., The Proteomics Identifications (PRIDE) database and associated tools: Status in 2013, Nucleic Acids Res, vol.41, pp.1063-1069, 2013.

M. Martin, Cutadapt removes adapter sequences from high-throughput sequencing reads, EMBnet. J, vol.17, pp.10-12, 2011.

A. M. Bolger, M. Lohse, and B. Usadel, Trimmomatic: A flexible trimmer for Illumina sequence data, Bioinformatics, vol.30, pp.2114-2120, 2014.

A. Bankevich, S. Nurk, D. Antipov, A. A. Gurevich, M. Dvorkin et al., SPAdes: A New Genome Assembly Algorithm and Its Applications to Single-Cell Sequencing, J. Comput. Biol, vol.19, pp.455-477, 2012.

D. Hyatt, G. L. Chen, P. F. Locascio, M. L. Land, F. W. Larimer et al., Prodigal: Prokaryotic gene recognition and translation initiation site identification, BMC Bioinform, vol.11, 2010.

T. Rognes, T. Flouri, B. Nichols, C. Quince, and F. Mahé, VSEARCH: A versatile open source tool for metagenomics, PeerJ, vol.18, 2016.

B. Langmead and S. L. Salzberg, Fast gapped-read alignment with Bowtie 2, Nat. Methods, vol.9, pp.357-359, 2012.

D. D. Kang, J. Froula, R. Egan, and Z. Wang, MetaBAT, an efficient tool for accurately reconstructing single genomes from complex microbial communities, PeerJ, vol.3, 1165.

D. H. Parks, M. Imelfort, C. T. Skennerton, P. Hugenholtz, G. W. Tyson et al., Assessing the quality of microbial genomes recovered from isolates, single cells, and metagenomes, Genome Res, vol.25, pp.1043-1055, 2015.

P. D. Schloss, S. L. Westcott, T. Ryabin, J. R. Hall, M. Hartmann et al., Introducing mothur: Open-source, platform-independent, community-supported software for describing and comparing microbial communities, Appl. Environ. Microbiol, vol.75, pp.7537-7541, 2009.

C. E. Shannon and W. Weaver, The mathematical theory of communication, Bell Syst. Tech. J, vol.34, pp.312-313, 1948.

O. Wang, G. M. Garrity, J. M. Tiedje, and J. R. Cole, Naïve Bayesian Classifier for Rapid Assignment of rRNA Sequences into the New Bacterial Taxonomy, Appl. Environ. Microbiol, vol.73, pp.5261-5267, 2007.

A. Tanca, A. Palomba, M. Deligios, T. Cubeddu, C. Fraumene et al., Evaluating the impact of different sequence databases on metaproteome analysis: Insights from a lab-assembled microbial mixture, PLoS ONE, vol.8, 2013.

T. Muth, C. A. Kolmeder, J. Salojärvi, S. Keskitalo, S. Varjosalo et al., Navigating through metaproteomics data: A logbook of database searching, Proteomics, vol.15, pp.3439-3453, 2015.

R. Hershberg, Mutation-The Engine of Evolution: Studying Mutation and Its Role in the Evolution of Bacteria, Cold Spring Harb. Perspect. Biol, 2015.

D. T. Troung, A. Tett, E. Pasolli, C. Huttenhower, and N. Segata, Microbial strain-level population structure and genetic diversity from metagenomes, Genome Res, vol.27, pp.626-638, 2017.

M. M. Savitski, M. Wilhelm, H. Hahne, B. Kuster, and M. Bantscheff, A Scalable Approach for Protein False Discovery Rate Estimation in Large Proteomic Data Sets, Mol. Cell. Proteom, vol.14, pp.2394-2404, 2015.

P. B. Eckburg, E. M. Bik, C. N. Bernstein, E. Purdom, L. Dethlefsen et al., Diversity of the human intestinal microbial flora, Science, vol.308, pp.1635-1638, 2005.

S. R. Gill, M. Pop, R. T. Deboy, P. B. Eckburg, P. J. Turnbaugh et al., Metagenomic Analysis of the Human Distal Gut Microbiome, Science, vol.312, pp.1355-1359, 2006.

X. Zhang, S. A. Deeke, Z. Ning, A. E. Starr, J. Butcher et al., Metaproteomics reveals associations between microbiome and intestinal extracellular vesicle proteins in pediatric inflammatory bowel disease, Nat. Commun, vol.9, 2018.

T. A. Cebula, Genetic and physiological modulation of anthracycline-induced mutagenesis in Salmonella typhimurium, Environ. Mutagen, vol.8, pp.675-692, 1986.

M. Riffle, D. H. May, E. Timmins-schiffman, M. P. Mikan, D. Jaschib et al., MetaGOmics: A Web-Based Tool for Peptide-Centric Functional and Taxonomic Analysis of Metaproteomics Data, Proteomes, vol.6, issue.2, 2018.

C. A. Kolmeder and W. S. De-vos, Metaproteomics of our microbiome-Developing insight in function and activity in man and model systems, J. Proteom, vol.97, pp.3-16, 2014.

J. E. Elias and S. P. Gygi, Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry, Nat. Methods, vol.604, pp.55-71, 2007.

G. Gonnelli, M. Stock, J. Verwaeren, D. Maddelein, B. De-baets et al., A decoy-free approach to the identification of peptides, J. Proteome Res, vol.14, pp.1792-1798, 2015.

H. Marx, S. Lemeer, S. Klaeger, T. Rattei, and B. Kuster, MScDB: A mass spectrometry-centric protein sequence database for proteomics, J. Proteome Res, vol.12, pp.2386-2398, 2013.

D. H. May, E. Timmins-schiffman, M. P. Mikan, H. R. Harvey, E. Borenstein et al., An alignment-free 'metapeptide' strategy for metaproteomic characterization of microbiome samples using shotgun metagenomic sequencing, J. Proteome Res, vol.15, pp.2697-2705, 2016.

P. Jagtap, J. Goslinga, J. A. Kooren, T. Mcgowan, M. S. Wroblewski et al., A two-step database search method improves sensitivity in peptide sequence matches for metaproteomics and proteogenomics studies, Proteomics, vol.13, pp.1352-1357, 2013.

D. P. Zolg, M. Wilhelm, K. Schnatbaum, J. Zerweck, T. Knaute et al., Building ProteomeTools based on a complete synthetic human proteome, Nat. Methods, vol.14, pp.259-262, 2017.

L. E. Papanicolas, D. L. Gordon, S. L. Wesselingh, and G. B. Rogers, Not Just Antibiotics: Is Cancer Chemotherapy Driving Antimicrobial Resistance?, Trends Microbiol, vol.26, pp.393-400, 2018.

W. Xiong, R. J. Giannone, M. J. Morowitz, J. F. Banfield, and R. L. Hettich, Development of an enhanced metaproteomic approach for deepening the microbiome characterization of the human infant gut, J. Proteome Res, vol.14, pp.133-141, 2014.

P. Y. Lee, S. F. Chin, H. M. Neoh, and R. Jamal, Metaproteomic analysis of human gut microbiota: Where are we heading?, J. Biomed. Sci, vol.24, 2017.

N. M. Griffin, J. Yu, F. Long, P. Oh, S. Shore et al., Label-free, normalized quantification of complex mass spectrometry data for proteomic analysis, Nat. Biotechnol, vol.28, pp.83-89, 2010.

D. H. Huson, S. Mitra, H. J. Ruscheweyh, N. Weber, and S. C. Schuster, Integrative analysis of environmental sequences using MEGAN4, Genome Res, vol.21, pp.1552-1560, 2011.

D. H. Huson and N. Weber, Microbial community analysis using MEGAN, Methods Enzymol, vol.531, pp.465-485, 2013.

J. Huerta-cepas, D. Szklarczyk, K. Forslund, H. Cook, D. Heller et al., 5: A hierarchical orthology framework with improved functional annotations for eukaryotic, prokaryotic and viral sequences, Nucleic Acids Res, vol.44, pp.286-293, 2016.

M. Kanehisa, Y. Sato, M. Kawashima, M. Furumichi, and M. Tanabe, KEGG as a reference resource for gene and protein annotation, Nucleic Acids Res, vol.44, pp.457-462, 2016.