Variation in the metagenomic analysis of fecal microbiome composition calls for a standardized operating approach
The reproducibility of human gut microbiome studies has been suboptimal across cohorts and study design choices. One possible reason for the disagreement is the introduction of systemic biases due to differences in methodologies. In our study, we utilized microbial metagenomic data sets from 2,722 f...
Saved in:
Published in | Microbiology spectrum Vol. 12; no. 12; p. e0151624 |
---|---|
Main Authors | , , , , , , , , , |
Format | Journal Article |
Language | English |
Published |
United States
American Society for Microbiology
30.10.2024
|
Subjects | |
Online Access | Get full text |
Cover
Loading…
Abstract | The reproducibility of human gut microbiome studies has been suboptimal across cohorts and study design choices. One possible reason for the disagreement is the introduction of systemic biases due to differences in methodologies. In our study, we utilized microbial metagenomic data sets from 2,722 fecal samples generated from a single research center to examine the extent to which sample storage and DNA extraction influence the quantification of microbial composition and compared this variable with other sources of technical and biological variation. Our research highlights the impact of DNA extraction methods when analyzing microbiome data and suggests that the microbiome profile may be influenced by differences in the extraction efficiency of bacterial species. With metagenomics sequencing being increasingly used in clinical biology, our findings provide insight into the challenges using metagenomics sequencing in clinical diagnostics, where the detection of certain species and its abundance relative to a “healthy reference” is key. |
---|---|
AbstractList | The reproducibility in microbiome studies is limited due to the lack of one gold-standard operating procedure. The aim of this study was to examine the impact of protocol variations on microbiome composition using metagenomic data sets from a single center. We assessed the variation in a data set consisted of 2,722 subjects, including 9 subcohorts harboring healthy subjects and patients with various disorders, such as inflammatory bowel disease, colorectal cancer, and type 2 diabetes. Two different DNA extraction kits, with or without lyticase, and two sample storage methods were compared. Our results indicated that DNA extraction had the largest impact on gut microbiota diversity among all host factors and sample operating procedures. Healthy subjects matched by age, body mass index, and sample operating methods exhibited reduced, yet significant differences (PERMANOVA, P < 0.05) in gut microbiota composition across studies. The variations contributed by DNA extraction were primarily driven by different recovery efficiency of gram-positive bacteria, e.g., phyla Firmicutes and Actinobacteria. This was further confirmed by a parallel comparison of fecal samples from five healthy subjects and a standard mock community. In addition, the DNA extraction method influenced DNA biomass, quality, and the detection of specific lineage-associated diseases. Sample operating approach and batch effects should be considered for cohorts with large sample size or longitudinal cohorts to ensure that source data were appropriately generated and analyzed. Comparison between samples processed with inconsistent methods should be dealt with caution. This study will promote the establishment of a sample operating standard to enhance our understanding of microbiome and translating in clinical practice.IMPORTANCEThe reproducibility of human gut microbiome studies has been suboptimal across cohorts and study design choices. One possible reason for the disagreement is the introduction of systemic biases due to differences in methodologies. In our study, we utilized microbial metagenomic data sets from 2,722 fecal samples generated from a single research center to examine the extent to which sample storage and DNA extraction influence the quantification of microbial composition and compared this variable with other sources of technical and biological variation. Our research highlights the impact of DNA extraction methods when analyzing microbiome data and suggests that the microbiome profile may be influenced by differences in the extraction efficiency of bacterial species. With metagenomics sequencing being increasingly used in clinical biology, our findings provide insight into the challenges using metagenomics sequencing in clinical diagnostics, where the detection of certain species and its abundance relative to a "healthy reference" is key.The reproducibility in microbiome studies is limited due to the lack of one gold-standard operating procedure. The aim of this study was to examine the impact of protocol variations on microbiome composition using metagenomic data sets from a single center. We assessed the variation in a data set consisted of 2,722 subjects, including 9 subcohorts harboring healthy subjects and patients with various disorders, such as inflammatory bowel disease, colorectal cancer, and type 2 diabetes. Two different DNA extraction kits, with or without lyticase, and two sample storage methods were compared. Our results indicated that DNA extraction had the largest impact on gut microbiota diversity among all host factors and sample operating procedures. Healthy subjects matched by age, body mass index, and sample operating methods exhibited reduced, yet significant differences (PERMANOVA, P < 0.05) in gut microbiota composition across studies. The variations contributed by DNA extraction were primarily driven by different recovery efficiency of gram-positive bacteria, e.g., phyla Firmicutes and Actinobacteria. This was further confirmed by a parallel comparison of fecal samples from five healthy subjects and a standard mock community. In addition, the DNA extraction method influenced DNA biomass, quality, and the detection of specific lineage-associated diseases. Sample operating approach and batch effects should be considered for cohorts with large sample size or longitudinal cohorts to ensure that source data were appropriately generated and analyzed. Comparison between samples processed with inconsistent methods should be dealt with caution. This study will promote the establishment of a sample operating standard to enhance our understanding of microbiome and translating in clinical practice.IMPORTANCEThe reproducibility of human gut microbiome studies has been suboptimal across cohorts and study design choices. One possible reason for the disagreement is the introduction of systemic biases due to differences in methodologies. In our study, we utilized microbial metagenomic data sets from 2,722 fecal samples generated from a single research center to examine the extent to which sample storage and DNA extraction influence the quantification of microbial composition and compared this variable with other sources of technical and biological variation. Our research highlights the impact of DNA extraction methods when analyzing microbiome data and suggests that the microbiome profile may be influenced by differences in the extraction efficiency of bacterial species. With metagenomics sequencing being increasingly used in clinical biology, our findings provide insight into the challenges using metagenomics sequencing in clinical diagnostics, where the detection of certain species and its abundance relative to a "healthy reference" is key. ABSTRACT The reproducibility in microbiome studies is limited due to the lack of one gold-standard operating procedure. The aim of this study was to examine the impact of protocol variations on microbiome composition using metagenomic data sets from a single center. We assessed the variation in a data set consisted of 2,722 subjects, including 9 subcohorts harboring healthy subjects and patients with various disorders, such as inflammatory bowel disease, colorectal cancer, and type 2 diabetes. Two different DNA extraction kits, with or without lyticase, and two sample storage methods were compared. Our results indicated that DNA extraction had the largest impact on gut microbiota diversity among all host factors and sample operating procedures. Healthy subjects matched by age, body mass index, and sample operating methods exhibited reduced, yet significant differences (PERMANOVA, P < 0.05) in gut microbiota composition across studies. The variations contributed by DNA extraction were primarily driven by different recovery efficiency of gram-positive bacteria, e.g., phyla Firmicutes and Actinobacteria. This was further confirmed by a parallel comparison of fecal samples from five healthy subjects and a standard mock community. In addition, the DNA extraction method influenced DNA biomass, quality, and the detection of specific lineage-associated diseases. Sample operating approach and batch effects should be considered for cohorts with large sample size or longitudinal cohorts to ensure that source data were appropriately generated and analyzed. Comparison between samples processed with inconsistent methods should be dealt with caution. This study will promote the establishment of a sample operating standard to enhance our understanding of microbiome and translating in clinical practice.IMPORTANCEThe reproducibility of human gut microbiome studies has been suboptimal across cohorts and study design choices. One possible reason for the disagreement is the introduction of systemic biases due to differences in methodologies. In our study, we utilized microbial metagenomic data sets from 2,722 fecal samples generated from a single research center to examine the extent to which sample storage and DNA extraction influence the quantification of microbial composition and compared this variable with other sources of technical and biological variation. Our research highlights the impact of DNA extraction methods when analyzing microbiome data and suggests that the microbiome profile may be influenced by differences in the extraction efficiency of bacterial species. With metagenomics sequencing being increasingly used in clinical biology, our findings provide insight into the challenges using metagenomics sequencing in clinical diagnostics, where the detection of certain species and its abundance relative to a “healthy reference” is key. The reproducibility in microbiome studies is limited due to the lack of one gold-standard operating procedure. The aim of this study was to examine the impact of protocol variations on microbiome composition using metagenomic data sets from a single center. We assessed the variation in a data set consisted of 2,722 subjects, including 9 subcohorts harboring healthy subjects and patients with various disorders, such as inflammatory bowel disease, colorectal cancer, and type 2 diabetes. Two different DNA extraction kits, with or without lyticase, and two sample storage methods were compared. Our results indicated that DNA extraction had the largest impact on gut microbiota diversity among all host factors and sample operating procedures. Healthy subjects matched by age, body mass index, and sample operating methods exhibited reduced, yet significant differences (PERMANOVA, P < 0.05) in gut microbiota composition across studies. The variations contributed by DNA extraction were primarily driven by different recovery efficiency of gram-positive bacteria, e.g., phyla Firmicutes and Actinobacteria. This was further confirmed by a parallel comparison of fecal samples from five healthy subjects and a standard mock community. In addition, the DNA extraction method influenced DNA biomass, quality, and the detection of specific lineage-associated diseases. Sample operating approach and batch effects should be considered for cohorts with large sample size or longitudinal cohorts to ensure that source data were appropriately generated and analyzed. Comparison between samples processed with inconsistent methods should be dealt with caution. This study will promote the establishment of a sample operating standard to enhance our understanding of microbiome and translating in clinical practice. The reproducibility of human gut microbiome studies has been suboptimal across cohorts and study design choices. One possible reason for the disagreement is the introduction of systemic biases due to differences in methodologies. In our study, we utilized microbial metagenomic data sets from 2,722 fecal samples generated from a single research center to examine the extent to which sample storage and DNA extraction influence the quantification of microbial composition and compared this variable with other sources of technical and biological variation. Our research highlights the impact of DNA extraction methods when analyzing microbiome data and suggests that the microbiome profile may be influenced by differences in the extraction efficiency of bacterial species. With metagenomics sequencing being increasingly used in clinical biology, our findings provide insight into the challenges using metagenomics sequencing in clinical diagnostics, where the detection of certain species and its abundance relative to a “healthy reference” is key. The reproducibility in microbiome studies is limited due to the lack of one gold-standard operating procedure. The aim of this study was to examine the impact of protocol variations on microbiome composition using metagenomic data sets from a single center. We assessed the variation in a data set consisted of 2,722 subjects, including 9 subcohorts harboring healthy subjects and patients with various disorders, such as inflammatory bowel disease, colorectal cancer, and type 2 diabetes. Two different DNA extraction kits, with or without lyticase, and two sample storage methods were compared. Our results indicated that DNA extraction had the largest impact on gut microbiota diversity among all host factors and sample operating procedures. Healthy subjects matched by age, body mass index, and sample operating methods exhibited reduced, yet significant differences (PERMANOVA, < 0.05) in gut microbiota composition across studies. The variations contributed by DNA extraction were primarily driven by different recovery efficiency of gram-positive bacteria, e.g., phyla Firmicutes and Actinobacteria. This was further confirmed by a parallel comparison of fecal samples from five healthy subjects and a standard mock community. In addition, the DNA extraction method influenced DNA biomass, quality, and the detection of specific lineage-associated diseases. Sample operating approach and batch effects should be considered for cohorts with large sample size or longitudinal cohorts to ensure that source data were appropriately generated and analyzed. Comparison between samples processed with inconsistent methods should be dealt with caution. This study will promote the establishment of a sample operating standard to enhance our understanding of microbiome and translating in clinical practice.IMPORTANCEThe reproducibility of human gut microbiome studies has been suboptimal across cohorts and study design choices. One possible reason for the disagreement is the introduction of systemic biases due to differences in methodologies. In our study, we utilized microbial metagenomic data sets from 2,722 fecal samples generated from a single research center to examine the extent to which sample storage and DNA extraction influence the quantification of microbial composition and compared this variable with other sources of technical and biological variation. Our research highlights the impact of DNA extraction methods when analyzing microbiome data and suggests that the microbiome profile may be influenced by differences in the extraction efficiency of bacterial species. With metagenomics sequencing being increasingly used in clinical biology, our findings provide insight into the challenges using metagenomics sequencing in clinical diagnostics, where the detection of certain species and its abundance relative to a "healthy reference" is key. The reproducibility in microbiome studies is limited due to the lack of one gold-standard operating procedure. The aim of this study was to examine the impact of protocol variations on microbiome composition using metagenomic data sets from a single center. We assessed the variation in a data set consisted of 2,722 subjects, including 9 subcohorts harboring healthy subjects and patients with various disorders, such as inflammatory bowel disease, colorectal cancer, and type 2 diabetes. Two different DNA extraction kits, with or without lyticase, and two sample storage methods were compared. Our results indicated that DNA extraction had the largest impact on gut microbiota diversity among all host factors and sample operating procedures. Healthy subjects matched by age, body mass index, and sample operating methods exhibited reduced, yet significant differences (PERMANOVA, P < 0.05) in gut microbiota composition across studies. The variations contributed by DNA extraction were primarily driven by different recovery efficiency of gram-positive bacteria, e.g., phyla Firmicutes and Actinobacteria. This was further confirmed by a parallel comparison of fecal samples from five healthy subjects and a standard mock community. In addition, the DNA extraction method influenced DNA biomass, quality, and the detection of specific lineage-associated diseases. Sample operating approach and batch effects should be considered for cohorts with large sample size or longitudinal cohorts to ensure that source data were appropriately generated and analyzed. Comparison between samples processed with inconsistent methods should be dealt with caution. This study will promote the establishment of a sample operating standard to enhance our understanding of microbiome and translating in clinical practice.IMPORTANCEThe reproducibility of human gut microbiome studies has been suboptimal across cohorts and study design choices. One possible reason for the disagreement is the introduction of systemic biases due to differences in methodologies. In our study, we utilized microbial metagenomic data sets from 2,722 fecal samples generated from a single research center to examine the extent to which sample storage and DNA extraction influence the quantification of microbial composition and compared this variable with other sources of technical and biological variation. Our research highlights the impact of DNA extraction methods when analyzing microbiome data and suggests that the microbiome profile may be influenced by differences in the extraction efficiency of bacterial species. With metagenomics sequencing being increasingly used in clinical biology, our findings provide insight into the challenges using metagenomics sequencing in clinical diagnostics, where the detection of certain species and its abundance relative to a “healthy reference” is key. |
Author | Xu, Zhilu Kamm, Michael A. Yeoh, Yun Kit Tun, Hein M. Fei, Na Zhang, Jingwan Ng, Siew C. Morrison, Mark Yu, Jun Chan, Francis Ka Leung |
Author_xml | – sequence: 1 givenname: Zhilu orcidid: 0000-0002-5552-7534 surname: Xu fullname: Xu, Zhilu – sequence: 2 givenname: Yun Kit surname: Yeoh fullname: Yeoh, Yun Kit – sequence: 3 givenname: Hein M. surname: Tun fullname: Tun, Hein M. – sequence: 4 givenname: Na surname: Fei fullname: Fei, Na – sequence: 5 givenname: Jingwan surname: Zhang fullname: Zhang, Jingwan – sequence: 6 givenname: Mark surname: Morrison fullname: Morrison, Mark – sequence: 7 givenname: Michael A. surname: Kamm fullname: Kamm, Michael A. – sequence: 8 givenname: Jun surname: Yu fullname: Yu, Jun – sequence: 9 givenname: Francis Ka Leung surname: Chan fullname: Chan, Francis Ka Leung – sequence: 10 givenname: Siew C. orcidid: 0000-0002-6850-4454 surname: Ng fullname: Ng, Siew C. |
BackLink | https://www.ncbi.nlm.nih.gov/pubmed/39475247$$D View this record in MEDLINE/PubMed |
BookMark | eNp1kU1v1DAQhiNUREvpD-CCfOSSJeOPeHNCqOKjUiUuwNUaO-NdrxI7tbNI7a8n3W2r9sDJ1szrZ8Z63lYnMUWqqvfQrAD4-lOZyM15P64aUNDWXL6qzji0qm5kp0-e3U-ri1J2TdMANIor_qY6FZ3Uikt9Vt38wRxwDimyENm8JTbSjBuKaQyOYcThtoTCkmeeHA5sqeZkQxqJuTROqYTD26U1FOZTZsjKjLHH3Ic76lmaKC_4uGE4TTmh276rXnscCl08nOfV729ff13-qK9_fr-6_HJdo1R6rrtWOFIWCLTnaysbLshqD2gBvLSyp16DE9Y7xQG04N5ZKahrVKd7zaU4r66O3D7hzkw5jJhvTcJgDoWUNwbzHNxAhou1AGo71_VCtkpbuRaq92qNy3iL3cL6fGRNeztS7yjOGYcX0JedGLZmk_4agBY6ofhC-PhAyOlmT2U2YyiOhgEjpX0xAjhvhRL8Pro6RrGM3OzSPi8WioHG3Hs3j97Nwbs5_PTD8-2e1nq0vATgGFjclZLJP0X-D_0H4IO_zA |
Cites_doi | 10.1136/gutjnl-2019-319635 10.1038/s41598-023-33959-6 10.1136/gutjnl-2020-323020 10.1128/mSystems.00392-19 10.1136/gutjnl-2020-324015 10.1038/s41564-021-01050-3 10.1053/j.gastro.2021.06.056 10.1007/s00248-010-9771-x 10.1158/1078-0432.CCR-16-1599 10.1053/j.gastro.2019.06.048 10.1038/nmeth.3589 10.1128/AEM.02627-17 10.1038/nbt.3981 10.1038/s41591-021-01552-x 10.1038/nbt.3960 10.1136/gutjnl-2019-318532 10.1186/s12866-020-01894-5 10.1111/j.1654-1103.2003.tb02228.x 10.1038/s41564-023-01426-7 10.1186/s40168-021-01059-0 10.1038/s41575-020-0341-5 10.1038/s41598-018-24280-8 10.1371/journal.pone.0202858 10.1038/d41586-018-01023-3 10.1016/s0167-7012(02)00018-0 10.1186/2049-2618-2-19 10.1038/s41591-019-0406-6 10.21105/joss.01686 10.1016/j.chom.2020.08.005 10.1093/bioinformatics/btu170 10.1053/j.gastro.2020.09.056 10.1038/s41582-022-00681-2 |
ContentType | Journal Article |
Copyright | Copyright © 2024 Xu et al. Copyright © 2024 Xu et al. 2024 Xu et al. |
Copyright_xml | – notice: Copyright © 2024 Xu et al. – notice: Copyright © 2024 Xu et al. 2024 Xu et al. |
DBID | AAYXX CITATION NPM 7X8 5PM DOA |
DOI | 10.1128/spectrum.01516-24 |
DatabaseName | CrossRef PubMed MEDLINE - Academic PubMed Central (Full Participant titles) DOAJ Directory of Open Access Journals |
DatabaseTitle | CrossRef PubMed MEDLINE - Academic |
DatabaseTitleList | MEDLINE - Academic CrossRef PubMed |
Database_xml | – sequence: 1 dbid: DOA name: DOAJ Directory of Open Access Journals url: https://www.doaj.org/ sourceTypes: Open Website – sequence: 2 dbid: NPM name: PubMed url: https://proxy.k.utb.cz/login?url=http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=PubMed sourceTypes: Index Database |
DeliveryMethod | fulltext_linktorsrc |
Discipline | Biology |
EISSN | 2165-0497 |
Editor | Claesen, Jan |
Editor_xml | – sequence: 1 givenname: Jan surname: Claesen fullname: Claesen, Jan |
ExternalDocumentID | oai_doaj_org_article_23831e69c9d34657b4835df58a1e1ba9 PMC11619352 spectrum01516-24 39475247 10_1128_spectrum_01516_24 |
Genre | Journal Article |
GrantInformation_xml | – fundername: InnoHK sequence: 0 – fundername: ; |
GroupedDBID | 53G AAGFI AAUOK AAYXX ADBBV AGVNZ ALMA_UNASSIGNED_HOLDINGS CITATION EJD FF~ FRP GROUPED_DOAJ H13 M~E OK1 RPM RSF NPM UCJ 7X8 5PM |
ID | FETCH-LOGICAL-a457t-963ce5b1e17f28b4023eb7f1ab11f4b4ded71c3bfc5211732fcb43e90597d7243 |
IEDL.DBID | AAUOK |
ISSN | 2165-0497 |
IngestDate | Wed Aug 27 01:30:37 EDT 2025 Thu Aug 21 18:29:18 EDT 2025 Fri Jul 11 06:55:55 EDT 2025 Thu Dec 05 18:27:12 EST 2024 Mon Jul 21 05:35:31 EDT 2025 Tue Jul 01 00:43:01 EDT 2025 |
IsDoiOpenAccess | true |
IsOpenAccess | true |
IsPeerReviewed | true |
IsScholarly | true |
Issue | 12 |
Keywords | batch effect DNA extraction microbiome metagenomics |
Language | English |
License | This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license. https://creativecommons.org/licenses/by/4.0 This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license. |
LinkModel | DirectLink |
MergedId | FETCHMERGED-LOGICAL-a457t-963ce5b1e17f28b4023eb7f1ab11f4b4ded71c3bfc5211732fcb43e90597d7243 |
Notes | ObjectType-Article-1 SourceType-Scholarly Journals-1 ObjectType-Feature-2 content type line 23 F.K.L.C. is a Board Member of CUHK Medical Centre. He is a co-founder, non-executive Board Chairman, and shareholder of GenieBiome Ltd. He receives patent royalties through his affiliated institutions. He has received fees as an advisor and honoraria as a speaker for Eisai Co. Ltd., AstraZeneca, Pfizer Inc., Takeda Pharmaceutical Co., and Takeda (China) Holdings Co. Ltd. S.C.N. has served as an advisory board member for Pfizer, Ferring, Janssen, and Abbvie and received honoraria as a speaker for Ferring, Tillotts, Menarini, Janssen, Abbvie, and Takeda. S.C.N. has received research grants through her affiliated institutions from Olympus, Ferring, and Abbvie. S.C.N. is a scientific co-founder and shareholder of GenieBiome Ltd. S.C.N. receives patent royalties through her affiliated institutions. Z.X., Y.K.Y., H.M.T., J.Z., F.K.L.C., and S.C.N. are named inventors of patent applications held by the CUHK and MagIC that cover the therapeutic and diagnostic use of microbiome. |
ORCID | 0000-0002-6850-4454 0000-0002-5552-7534 |
OpenAccessLink | https://journals.asm.org/doi/10.1128/spectrum.01516-24 |
PMID | 39475247 |
PQID | 3122635322 |
PQPubID | 23479 |
PageCount | 13 |
ParticipantIDs | doaj_primary_oai_doaj_org_article_23831e69c9d34657b4835df58a1e1ba9 pubmedcentral_primary_oai_pubmedcentral_nih_gov_11619352 proquest_miscellaneous_3122635322 asm2_journals_10_1128_spectrum_01516_24 pubmed_primary_39475247 crossref_primary_10_1128_spectrum_01516_24 |
ProviderPackageCode | CITATION AAYXX |
PublicationCentury | 2000 |
PublicationDate | 20241030 |
PublicationDateYYYYMMDD | 2024-10-30 |
PublicationDate_xml | – month: 10 year: 2024 text: 20241030 day: 30 |
PublicationDecade | 2020 |
PublicationPlace | United States |
PublicationPlace_xml | – name: United States – name: 1752 N St., N.W., Washington, DC |
PublicationTitle | Microbiology spectrum |
PublicationTitleAbbrev | Spectrum |
PublicationTitleAlternate | Microbiol Spectr |
PublicationYear | 2024 |
Publisher | American Society for Microbiology |
Publisher_xml | – sequence: 0 name: American Society for Microbiology – name: American Society for Microbiology |
References | e_1_3_5_28_2 e_1_3_5_27_2 e_1_3_5_26_2 e_1_3_5_24_2 e_1_3_5_23_2 e_1_3_5_22_2 e_1_3_5_21_2 e_1_3_5_29_2 e_1_3_5_2_2 e_1_3_5_8_2 e_1_3_5_20_2 e_1_3_5_7_2 e_1_3_5_9_2 e_1_3_5_4_2 e_1_3_5_3_2 e_1_3_5_6_2 e_1_3_5_5_2 e_1_3_5_17_2 e_1_3_5_16_2 e_1_3_5_15_2 e_1_3_5_14_2 e_1_3_5_12_2 e_1_3_5_35_2 e_1_3_5_13_2 e_1_3_5_34_2 e_1_3_5_10_2 e_1_3_5_33_2 e_1_3_5_11_2 e_1_3_5_32_2 e_1_3_5_19_2 e_1_3_5_18_2 McMurdie PJ (e_1_3_5_25_2) 2012; 2012 e_1_3_5_31_2 e_1_3_5_30_2 Lim, MY, Park, Y-S, Kim, J-H, Nam, Y-D (B11) 2020; 20 B23 Dixon, P (B25) 2003; 14 Bolger, AM, Lohse, M, Usadel, B (B19) 2014; 30 Sakowski, E, Uritskiy, G, Cooper, R, Gomes, M, McLaren, MR, Meisel, JS, Mickol, RL, Mintz, CD, Mongodin, EF, Pop, M (B9) 2019; 4 Gupta, A, Osadchiy, V, Mayer, EA (B4) 2020; 17 Wirbel, J, Pyl, PT, Kartal, E, Zych, K, Kashani, A, Milanese, A, Fleck, JS, Voigt, AY, Palleja, A, Ponnudurai, R (B7) 2019; 25 Zuo, T, Sun, Y, Wan, Y, Yeoh, YK, Zhang, F, Cheung, CP, Chen, N, Luo, J, Wang, W, Sung, JJY, Chan, PKS, Wang, K, Chan, FKL, Miao, Y, Ng, SC (B16) 2020; 28 Nearing, JT, Comeau, AM, Langille, MGI (B8) 2021; 9 Liang, Q, Chiu, J, Chen, Y, Huang, Y, Higashimori, A, Fang, J, Brim, H, Ashktorab, H, Ng, SC, Ng, SSM, Zheng, S, Chan, FKL, Sung, JJY, Yu, J (B18) 2017; 23 Munafò, MR, Davey Smith, G (B32) 2018; 553 Ó Cuív, P, Aguirre de Cárcer, D, Jones, M, Klaassens, ES, Worthley, DL, Whitehall, VLJ, Kang, S, McSweeney, CS, Leggett, BA, Morrison, M (B28) 2011; 61 Walker, AW, Hoyles, L (B33) 2023; 8 Costea, PI, Zeller, G, Sunagawa, S, Pelletier, E, Alberti, A, Levenez, F, Tramontano, M, Driessen, M, Hercog, R, Jung, F-E (B6) 2017; 35 McGaughey, KD, Yilmaz-Swenson, T, Elsayed, NM, Cruz, DA, Rodriguez, RR, Kritzer, MD, Peterchev, AV, Gray, M, Lewis, SR, Roach, J, Wetsel, WC, Williamson, DE (B29) 2018; 13 Elie, C, Perret, M, Hage, H, Sentausa, E, Hesketh, A, Louis, K, Fritah-Lafont, A, Leissner, P, Vachon, C, Rostaing, H, Reynier, F, Gervasi, G, Saliou, A (B31) 2023; 13 Tan, AH, Lim, SY, Lang, AE (B3) 2022; 18 Wan, Y, Zuo, T, Xu, Z, Zhang, F, Zhan, H, Chan, D, Leung, T-F, Yeoh, YK, Chan, FKL, Chan, R, Ng, SC (B15) 2022; 71 Mirzayi, C, Renson, A, Zohra, F, Elsafoury, S, Geistlinger, L, Kasselman, LJ, Eckenrode, K, van de Wijgert, J (B34) 2021; 27 Yang, K, Niu, J, Zuo, T, Sun, Y, Xu, Z, Tang, W, Liu, Q, Zhang, J, Ng, EKW, Wong, SKH, Yeoh, YK, Chan, PKS, Chan, FKL, Miao, Y, Ng, SC (B14) 2021; 161 Wesolowska-Andersen, A, Bahl, MI, Carvalho, V, Kristiansen, K, Sicheritz-Pontén, T, Gupta, R, Licht, TR (B30) 2014; 2 Lee, M, Chang, EB (B2) 2021; 160 Pollock, J, Glendinning, L, Wisedchanwet, T, Watson, M (B26) 2018; 84 Mills, RH, Dulai, PS, Vázquez-Baeza, Y, Sauceda, C, Daniel, N, Gerner, RR, Batachari, LE, Malfavon, M, Zhu, Q, Weldon, K, Humphrey, G, Carrillo-Terrazas, M, Goldasich, LD, Bryant, M, Raffatellu, M, Quinn, RA, Gewirtz, AT, Chassaing, B, Chu, H, Sandborn, WJ, Dorrestein, PC, Knight, R, Gonzalez, DJ (B5) 2022; 7 Yeoh, YK, Zuo, T, Lui, G-Y, Zhang, F, Liu, Q, Li, AY, Chung, AC, Cheung, CP, Tso, EY, Fung, KS, Chan, V, Ling, L, Joynt, G, Hui, D-C, Chow, KM, Ng, SSS, Li, T-M, Ng, RW, Yip, TC, Wong, G-H, Chan, FK, Wong, CK, Chan, PK, Ng, SC (B17) 2021; 70 Song, M, Chan, AT, Sun, J (B1) 2020; 158 McOrist, AL, Jackson, M, Bird, AR (B10) 2002; 50 Yeoh, YK, Chen, Z, Wong, MCS, Hui, M, Yu, J, Ng, SC, Sung, JJY, Chan, FKL, Chan, PKS (B13) 2020; 69 McMurdie, PJ, Holmes, S (B24) 2012; 2012 Sinha, R, Abu-Ali, G, Vogtmann, E, Fodor, AA, Ren, B, Amir, A, Schwager, E, Crabtree, J, Ma, S, Abnet, CC, Knight, R, White, O, Huttenhower, C (B27) 2017; 35 Liang, JQ, Li, T, Nakatsu, G, Chen, Y-X, Yau, TO, Chu, E, Wong, S, Szeto, CH, Ng, SC, Chan, FKL, Fang, J-Y, Sung, JJY, Yu, J (B12) 2020; 69 Zaheer, R, Noyes, N, Ortega Polo, R, Cook, SR, Marinier, E, Van Domselaar, G, Belk, KE, Morley, PS, McAllister, TA (B20) 2018; 8 Truong, DT, Franzosa, EA, Tickle, TL, Scholz, M, Weingart, G, Pasolli, E, Tett, A, Huttenhower, C, Segata, N (B21) 2015; 12 Wickham, H, Averick, M, Bryan, J, Chang, W, McGowan, L, François, R, Grolemund, G, Hayes, A, Henry, L, Hester, J, Kuhn, M, Pedersen, T, Miller, E, Bache, S, Müller, K, Ooms, J, Robinson, D, Seidel, D, Spinu, V, Takahashi, K, Vaughan, D, Wilke, C, Woo, K, Yutani, H (B22) 2019; 4 |
References_xml | – ident: e_1_3_5_14_2 doi: 10.1136/gutjnl-2019-319635 – ident: e_1_3_5_32_2 doi: 10.1038/s41598-023-33959-6 – ident: e_1_3_5_18_2 doi: 10.1136/gutjnl-2020-323020 – ident: e_1_3_5_10_2 doi: 10.1128/mSystems.00392-19 – ident: e_1_3_5_16_2 doi: 10.1136/gutjnl-2020-324015 – ident: e_1_3_5_6_2 doi: 10.1038/s41564-021-01050-3 – ident: e_1_3_5_15_2 doi: 10.1053/j.gastro.2021.06.056 – ident: e_1_3_5_29_2 doi: 10.1007/s00248-010-9771-x – ident: e_1_3_5_19_2 doi: 10.1158/1078-0432.CCR-16-1599 – ident: e_1_3_5_2_2 doi: 10.1053/j.gastro.2019.06.048 – ident: e_1_3_5_22_2 doi: 10.1038/nmeth.3589 – ident: e_1_3_5_27_2 doi: 10.1128/AEM.02627-17 – volume: 2012 start-page: 235 year: 2012 ident: e_1_3_5_25_2 article-title: Phyloseq: a bioconductor package for handling and analysis of high-throughput phylogenetic sequence data publication-title: Pac Symp Biocomput – ident: e_1_3_5_28_2 doi: 10.1038/nbt.3981 – ident: e_1_3_5_35_2 doi: 10.1038/s41591-021-01552-x – ident: e_1_3_5_7_2 doi: 10.1038/nbt.3960 – ident: e_1_3_5_13_2 doi: 10.1136/gutjnl-2019-318532 – ident: e_1_3_5_12_2 doi: 10.1186/s12866-020-01894-5 – ident: e_1_3_5_24_2 – ident: e_1_3_5_26_2 doi: 10.1111/j.1654-1103.2003.tb02228.x – ident: e_1_3_5_34_2 doi: 10.1038/s41564-023-01426-7 – ident: e_1_3_5_9_2 doi: 10.1186/s40168-021-01059-0 – ident: e_1_3_5_5_2 doi: 10.1038/s41575-020-0341-5 – ident: e_1_3_5_21_2 doi: 10.1038/s41598-018-24280-8 – ident: e_1_3_5_30_2 doi: 10.1371/journal.pone.0202858 – ident: e_1_3_5_33_2 doi: 10.1038/d41586-018-01023-3 – ident: e_1_3_5_11_2 doi: 10.1016/s0167-7012(02)00018-0 – ident: e_1_3_5_31_2 doi: 10.1186/2049-2618-2-19 – ident: e_1_3_5_8_2 doi: 10.1038/s41591-019-0406-6 – ident: e_1_3_5_23_2 doi: 10.21105/joss.01686 – ident: e_1_3_5_17_2 doi: 10.1016/j.chom.2020.08.005 – ident: e_1_3_5_20_2 doi: 10.1093/bioinformatics/btu170 – ident: e_1_3_5_3_2 doi: 10.1053/j.gastro.2020.09.056 – ident: e_1_3_5_4_2 doi: 10.1038/s41582-022-00681-2 – volume: 35 start-page: 1077 year: 2017 end-page: 1086 ident: B27 article-title: Assessment of variation in microbial community amplicon sequencing by the microbiome quality control (MBQC) project consortium publication-title: Nat Biotechnol doi: 10.1038/nbt.3981 – volume: 69 start-page: 1998 year: 2020 end-page: 2007 ident: B13 article-title: Southern Chinese populations harbour non-nucleatum Fusobacteria possessing homologues of the colorectal cancer-associated FadA virulence factor publication-title: Gut doi: 10.1136/gutjnl-2019-319635 – volume: 161 start-page: 1257 year: 2021 end-page: 1269 ident: B14 article-title: Alterations in the gut virome in obesity and type 2 diabetes mellitus publication-title: Gastroenterology doi: 10.1053/j.gastro.2021.06.056 – ident: B23 article-title: ggpubr WH . ggpubr: ‘ggplot2’ based publication ready plots – volume: 25 start-page: 679 year: 2019 end-page: 689 ident: B7 article-title: Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer publication-title: Nat Med doi: 10.1038/s41591-019-0406-6 – volume: 20 year: 2020 ident: B11 article-title: Evaluation of fecal DNA extraction protocols for human gut microbiome studies publication-title: BMC Microbiol doi: 10.1186/s12866-020-01894-5 – volume: 50 start-page: 131 year: 2002 end-page: 139 ident: B10 article-title: A comparison of five methods for extraction of bacterial DNA from human faecal samples publication-title: J Microbiol Methods doi: 10.1016/s0167-7012(02)00018-0 – volume: 2012 start-page: 235 year: 2012 end-page: 246 ident: B24 article-title: Phyloseq: a bioconductor package for handling and analysis of high-throughput phylogenetic sequence data publication-title: Pac Symp Biocomput – volume: 61 start-page: 353 year: 2011 end-page: 362 ident: B28 article-title: The effects from DNA extraction methods on the evaluation of microbial diversity associated with human colonic tissue publication-title: Microb Ecol doi: 10.1007/s00248-010-9771-x – volume: 28 start-page: 741 year: 2020 end-page: 751 ident: B16 article-title: Human-gut-DNA virome variations across geography, ethnicity, and urbanization publication-title: Cell Host Microbe doi: 10.1016/j.chom.2020.08.005 – volume: 13 year: 2018 ident: B29 article-title: Comparative evaluation of a new magnetic bead-based DNA extraction method from fecal samples for downstream next-generation 16S rRNA gene sequencing publication-title: PLoS One doi: 10.1371/journal.pone.0202858 – volume: 84 year: 2018 ident: B26 article-title: The madness of microbiome: attempting to find consensus "Best Practice" for 16S microbiome studies publication-title: Appl Environ Microbiol doi: 10.1128/AEM.02627-17 – volume: 7 start-page: 262 year: 2022 end-page: 276 ident: B5 article-title: Multi-omics analyses of the ulcerative colitis gut microbiome link Bacteroides vulgatus proteases with disease severity publication-title: Nat Microbiol doi: 10.1038/s41564-021-01050-3 – volume: 160 start-page: 524 year: 2021 end-page: 537 ident: B2 article-title: Inflammatory bowel diseases (IBD) and the microbiome-searching the crime scene for clues publication-title: Gastroenterology doi: 10.1053/j.gastro.2020.09.056 – volume: 23 start-page: 2061 year: 2017 end-page: 2070 ident: B18 article-title: Fecal bacteria act as novel biomarkers for noninvasive diagnosis of colorectal cancer publication-title: Clin Cancer Res doi: 10.1158/1078-0432.CCR-16-1599 – volume: 158 start-page: 322 year: 2020 end-page: 340 ident: B1 article-title: Influence of the gut microbiome, diet, and environment on risk of colorectal cancer publication-title: Gastroenterology doi: 10.1053/j.gastro.2019.06.048 – volume: 70 start-page: 698 year: 2021 end-page: 706 ident: B17 article-title: Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19 publication-title: Gut doi: 10.1136/gutjnl-2020-323020 – volume: 14 start-page: 927 year: 2003 end-page: 930 ident: B25 article-title: VEGAN, a package of R functions for community ecology publication-title: J Veg Sci doi: 10.1111/j.1654-1103.2003.tb02228.x – volume: 35 start-page: 1069 year: 2017 end-page: 1076 ident: B6 article-title: Towards standards for human fecal sample processing in metagenomic studies publication-title: Nat Biotechnol doi: 10.1038/nbt.3960 – volume: 27 start-page: 1885 year: 2021 end-page: 1892 ident: B34 article-title: Reporting guidelines for human microbiome research: the STORMS checklist publication-title: Nat Med doi: 10.1038/s41591-021-01552-x – volume: 17 start-page: 655 year: 2020 end-page: 672 ident: B4 article-title: Brain-gut-microbiome interactions in obesity and food addiction publication-title: Nat Rev Gastroenterol Hepatol doi: 10.1038/s41575-020-0341-5 – volume: 9 year: 2021 ident: B8 article-title: Identifying biases and their potential solutions in human microbiome studies publication-title: Microbiome doi: 10.1186/s40168-021-01059-0 – volume: 18 start-page: 476 year: 2022 end-page: 495 ident: B3 article-title: The microbiome-gut-brain axis in Parkinson disease - from basic research to the clinic publication-title: Nat Rev Neurol doi: 10.1038/s41582-022-00681-2 – volume: 12 start-page: 902 year: 2015 end-page: 903 ident: B21 article-title: MetaPhlAn2 for enhanced metagenomic taxonomic profiling publication-title: Nat Methods doi: 10.1038/nmeth.3589 – volume: 30 start-page: 2114 year: 2014 end-page: 2120 ident: B19 article-title: Trimmomatic: a flexible trimmer for Illumina sequence data publication-title: Bioinformatics doi: 10.1093/bioinformatics/btu170 – volume: 8 start-page: 1392 year: 2023 end-page: 1396 ident: B33 article-title: Human microbiome myths and misconceptions publication-title: Nat Microbiol doi: 10.1038/s41564-023-01426-7 – volume: 71 start-page: 910 year: 2022 end-page: 918 ident: B15 article-title: Underdevelopment of the gut microbiota and bacteria species as non-invasive markers of prediction in children with autism spectrum disorder publication-title: Gut doi: 10.1136/gutjnl-2020-324015 – volume: 4 start-page: 1686 year: 2019 ident: B22 article-title: Welcome to the tidyverse publication-title: JOSS doi: 10.21105/joss.01686 – volume: 69 start-page: 1248 year: 2020 end-page: 1257 ident: B12 article-title: A novel faecal Lachnoclostridium marker for the non-invasive diagnosis of colorectal adenoma and cancer publication-title: Gut doi: 10.1136/gutjnl-2019-318532 – volume: 8 year: 2018 ident: B20 article-title: Impact of sequencing depth on the characterization of the microbiome and resistome publication-title: Sci Rep doi: 10.1038/s41598-018-24280-8 – volume: 553 start-page: 399 year: 2018 end-page: 401 ident: B32 article-title: Robust research needs many lines of evidence publication-title: Nature New Biol doi: 10.1038/d41586-018-01023-3 – volume: 13 year: 2023 ident: B31 article-title: Comparison of DNA extraction methods for 16S rRNA gene sequencing in the analysis of the human gut microbiome publication-title: Sci Rep doi: 10.1038/s41598-023-33959-6 – volume: 4 year: 2019 ident: B9 article-title: Current state of and future opportunities for prediction in microbiome research: report from the mid-Atlantic microbiome meet-up in Baltimore on 9 January 2019 publication-title: mSystems doi: 10.1128/mSystems.00392-19 – volume: 2 start-page: 19 year: 2014 ident: B30 article-title: Choice of bacterial DNA extraction method from fecal material influences community structure as evaluated by metagenomic analysis publication-title: Microbiome doi: 10.1186/2049-2618-2-19 |
SSID | ssj0001105252 |
Score | 2.3245718 |
Snippet | The reproducibility of human gut microbiome studies has been suboptimal across cohorts and study design choices. One possible reason for the disagreement is... The reproducibility in microbiome studies is limited due to the lack of one gold-standard operating procedure. The aim of this study was to examine the impact... ABSTRACT The reproducibility in microbiome studies is limited due to the lack of one gold-standard operating procedure. The aim of this study was to examine... |
SourceID | doaj pubmedcentral proquest asm2 pubmed crossref |
SourceType | Open Website Open Access Repository Aggregation Database Index Database |
StartPage | e0151624 |
SubjectTerms | batch effect DNA extraction Human Microbiome metagenomics microbiome Research Article |
SummonAdditionalLinks | – databaseName: DOAJ Directory of Open Access Journals dbid: DOA link: http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwrV3NaxQxFA-yIPQirVY7WiUFQRDGbpKXycyxiqUI9eRKbyGfdKAz27rbg_71fcnMLLsievE6CUx4H3m_l-T9HiFvawjAHIul4I3FBMU0ZS3reakQ70vHuDdVqne-_FpdLODLlbzaavWV3oQN9MCD4E4xpAgWqsY1XkAllQXEDD7K2rDArMmlexjztpKpfLrCUn82Pl5j4h58mgsXf9x3HzD-sapMFe4zs-r4TjzKtP1_wpq_P5ncikHn--TJCB7p2bDoA_Io9E_J46Gd5M9n5O47Jr5Z0rTtKSI72oW1SSysXeuoGelH6DLSGFA1tGsHFqYu0PSyfHy-RXHoZkURzFJDp5OG9lfwdHmbGJgx1tGJifyQLM4_f_t0UY4tFUoDUq1LdDcXpEXRqchri8mjCFZFZixjESz44BVzwkaHYZ0pwaOzIEKDIEx5xUE8J7N-2YcjQsFEz90c0yHhwSPukE7gbgUAEjNdVxXkXZKvHn1ipXO6wWs9aUJnTWgOBXk_qUDfDhwbf5v8MSlpMzHRY-cPaDR6NBr9L6MpyMmkYo3ulO5ITB-W9ystGE_0PLjNFeTFoPLNr0QDSnJQBal3jGFnLbsjfXudKbsZAusGse7L_7H6V2SPI7TKEXR-TGYon_AaodHavsle8AC0Kg0f priority: 102 providerName: Directory of Open Access Journals |
Title | Variation in the metagenomic analysis of fecal microbiome composition calls for a standardized operating approach |
URI | https://www.ncbi.nlm.nih.gov/pubmed/39475247 https://journals.asm.org/doi/10.1128/spectrum.01516-24 https://www.proquest.com/docview/3122635322 https://pubmed.ncbi.nlm.nih.gov/PMC11619352 https://doaj.org/article/23831e69c9d34657b4835df58a1e1ba9 |
Volume | 12 |
hasFullText | 1 |
inHoldings | 1 |
isFullTextHit | |
isPrint | |
link | http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwjV1Lb9QwELZKKyQuiPJMgcpISEhIKWt7HCfHBbWqQMCFRb1ZfopITbaw20P59YydZGFRVfUaO3Iyn8fzjR-fCXldQwDmWCwFbywmKKYpa1nPSoV8XzrGvanSeefPX6rTBXw8k2c7pJrOwowWXB2ZVZcX8jeezet3-fDhr8vuCGMYq0oOd8ie5A2gQ-7N54uvn_7OrrB0PxsflzGvfRfHYGyDb8WjLNt_Hdf8f8vkPzHo5AG5P5JHOh_Q3ic7oX9I7g7XSV49Ij-_Y-KbLU3bniKzo11Ym6TC2rWOmlF-hC4jjQGhoV07qDB1gaad5eP2LYpF5yuKZJYaOs00tL-Dp8uLpMCMsY5OSuSPyeLk-NuH03K8UqE0INW6RHdzQVoWmIq8tpg8imBVZMYyFsGCD14xJ2x0GNaZEjw6CyI0SMKUVxzEE7LbL_vwjFAw0XM3w3RIePDIO6QTOFoBgMRM11UFeZPsqydEdU43eK0nJHRGQnMoyNsJAn0xaGzcVPl9AmlTMclj5wfYWfTobRp5iGChalzjBVRSWUCi6aOsDf65NU1BXk0Qa3SntEZi-rC8XGnBeJLnwWGuIE8HyDdNiQaU5KAKUm91hq1v2S7p2x9ZspshsW6Q6x7c2ijPyT2O_CmHydkLsouF4SXyn7U9HDv7YZ4_-AN14QYh |
linkProvider | American Society for Microbiology |
linkToHtml | http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwjV1Lb9QwELagFYIL4k14GgkJCSll_YqT44KoFvrg0kW9WX6KoCbbNtsD_HrGjlNYVCGusSMnHo_nG8_4G4Re19xzYkkoGW0MOCi6KWtRz0oJeF9YQp2u4n3ng8NqseSfj8VxzqqMd2G-x7q8J8OOHroUx4-KHQ-icz3C-l26gHh-0e2AHSNVSfl1tB1jh-B4bc_nyy97v09YSKzRRnMo88p3YR-GgeiGTUrU_Vfhzb_TJv-wQ7t30O0MIPF8lPhddM3399CNsaTkj_vo7Cs4v2m2cdtjQHe482sdmVi71mKdKUjwKuDgQTy4a0cmps7jmF2eU7gwNJ0MGAAt1ng6bWh_eodXp5GFGewdntjIH6Dl7sejD4syl1UoNRdyXYLKWS8M8UQGWhtwIJk3MhBtCAnccOedJJaZYMG0E8losIYz3wAQk05Szh6irX7V-8cIcx0ctTNwiZjjDrCHsAx2LM65AG_XVgV6E-dXZb0YVHI5aK0mSagkCUV5gd5OIlCnI8_Gvzq_j0K67BgpstMDWDEqa5wCLMKIrxrbOMYrIQ0HsOmCqDX8udFNgV5NIlagUjFOonu_uhgUIzRS9MBWV6BHo8gvh2INl4JyWaB6YzFsfMtmS99-S7TdBMB1A3j3yX9Pykt0c3F0sK_2Px3uPUW3KOCpZDZnz9AWdPTPAQ-tzYu88H8BrKQJjw |
linkToPdf | http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwjV1Jb9QwFLZgKhCXip2wGgkJCSnteIuT47QwKhQKBwb1ZnkVkUhmYKaH8ut5dpzCoApxjR05eZ-f3_e8fEboRc09J5aEktHGQIKim7IW9bSUwPeFJdTpKp53_nBSHS34u1NxmndVxrMw2YLrPb3u0kJ-9OyVC_k-wno_HUD8cdbtQRwjVUn5VbSTFqsmaGc2W3w8_j3DQuIdbTQvZV76LozD0A7diklJuv8yvvn3tsk_4tD8JtrNBBLPBsRvoSu-v42uDVdKnt9B379A8pusjdseA7vDnd_oqMTatRbrLEGClwEHD_Dgrh2UmDqP4-7yvIULQ9G3NQZCizUeZxvan97h5SqqMEO8w6Ma-V20mL_5fHhU5msVSs2F3JTgctYLQzyRgdYGEkjmjQxEG0ICN9x5J4llJlgI7UQyGqzhzDdAxKSTlLN7aNIve_8AYa6Do3YKKRFz3AH3EJbBiMU5F5Dt2qpAL6N91YiqSikHrdWIhEpIKMoL9GqEQK0GnY1_VT6IIF1UjBLZ6QF0GJU9TgEXYcRXjW0c45WQhgPZdEHUGv7c6KZAz0eIFbhUXCfRvV-erRUjNEr0wFBXoPsD5BdNsYZLQbksUL3VGba-Zbukb78m2W4C5LoBvvvwv43yDF3_9Hqu3r89OX6EblCgUylqTh-jCdTzT4AObczT3O9_AYaaCSs |
openUrl | ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fsummon.serialssolutions.com&rft_val_fmt=info%3Aofi%2Ffmt%3Akev%3Amtx%3Ajournal&rft.genre=article&rft.atitle=Variation+in+the+metagenomic+analysis+of+fecal+microbiome+composition+calls+for+a+standardized+operating+approach&rft.jtitle=Microbiology+spectrum&rft.au=Xu%2C+Zhilu&rft.au=Yeoh%2C+Yun+Kit&rft.au=Tun%2C+Hein+M.&rft.au=Fei%2C+Na&rft.date=2024-10-30&rft.pub=American+Society+for+Microbiology&rft.eissn=2165-0497&rft.volume=12&rft.issue=12&rft_id=info:doi/10.1128%2Fspectrum.01516-24&rft.externalDocID=spectrum01516-24 |
thumbnail_l | http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/lc.gif&issn=2165-0497&client=summon |
thumbnail_m | http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/mc.gif&issn=2165-0497&client=summon |
thumbnail_s | http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/sc.gif&issn=2165-0497&client=summon |