Supplementary Table 8 Spearman's Correlations Between Targeted Urinary Urolithins and Microbiota

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Table 8 Spearman's Correlations Between Targeted Urinary Urolithins and Microbiota Supplementary material Gut Supplementary Table 8 Spearman's correlations between targeted urinary urolithins and microbiota. Urolithin- Urolithin- Urolithin- Total A- B- C- Urolithins Family level Taxonomy glucuronid glucuronid glucuronid (A+B+C) e e e Actinobacteria; Actinobacteria; Bifidobacteriales; Bifidobacteriaceae; Bifidobacterium adolescentis_msp_0263 -0.18 -0.09 -0.16 -0.18 Bifidobacteriaceae Bifidobacterium; Bifidobacterium adolescentis Actinobacteria; Actinobacteria; Bifidobacteriales; Bifidobacteriaceae; Bifidobacterium bifidum_msp_0419 -0.12 -0.2 -0.08 -0.13 Bifidobacteriaceae Bifidobacterium; Bifidobacterium bifidum Actinobacteria; Coriobacteriia; Coriobacteriales; Coriobacteriaceae; Collinsella; Collinsella aerofaciens_msp_1244 -0.15 -0.06 -0.04 -0.18 Coriobacteriaceae Collinsella aerofaciens Actinobacteria; Coriobacteriia; Eggerthellales; Eggerthellaceae; Adlercreutzia; unclassified Adlercreutzia_msp_0396 0.09 0.01 0.16 0.12 Eggerthellaceae unclassified Adlercreutzia Actinobacteria; Coriobacteriia; Eggerthellales; Eggerthellaceae; Eggerthella; Eggerthella lenta_msp_0573 0.03 -0.15 0.08 0.03 Eggerthellaceae Eggerthella lenta Actinobacteria; Coriobacteriia; Eggerthellales; Eggerthellaceae; Gordonibacter; Gordonibacter urolithinfaciens_msp_1339 0.19 -0.05 0.18 0.19 Eggerthellaceae Gordonibacter urolithinfaciens Bacteroidetes; Bacteroidia; Bacteroidales; Bacteroidaceae; Bacteroides; Bacteroides cellulosilyticus_msp_0003 0.12 0.11 0.15 0.15 Bacteroidaceae Bacteroides cellulosilyticus Bacteroidetes; Bacteroidia; Bacteroidales; Bacteroidaceae; Bacteroides; Bacteroides dorei_msp_0035 0.12 0.23 0.14 0.19 Bacteroidaceae Bacteroides dorei Bacteroidetes; Bacteroidia; Bacteroidales; Bacteroidaceae; Bacteroides; Bacteroides faecis_msp_0019 0.09 -0.11 0.15 0.07 Bacteroidaceae Bacteroides faecis Bacteroidetes; Bacteroidia; Bacteroidales; Bacteroidaceae; Bacteroides; Bacteroides salyersiae_msp_0037 0.17 0.11 0.05 0.17 Bacteroidaceae Bacteroides salyersiae Bacteroidetes; Bacteroidia; Bacteroidales; Bacteroidaceae; Bacteroides; Bacteroides sp. CAG:144_msp_0412 0.2 0.26 0.15 0.24 Bacteroidaceae Bacteroides sp. CAG:144 Coprobacter secundus == Gabonia Bacteroidetes; Bacteroidia; Bacteroidales; Barnesiellaceae; Coprobacter; 0.2 0.13 0.14 0.2 Barnesiellaceae massiliensis_msp_0198 Coprobacter secundus == Gabonia massiliensis Alistipes inops == Tidjanibacter Bacteroidetes; Bacteroidia; Bacteroidales; Rikenellaceae; Alistipes; Alistipes 0.05 -0.17 0 -0.02 Rikenellaceae massiliensis_msp_0230 inops == Tidjanibacter massiliensis Bacteroidetes; Bacteroidia; Bacteroidales; Rikenellaceae; Alistipes; Alistipes Alistipes obesi_msp_0225 0.1 0.23 0.08 0.16 Rikenellaceae obesi Bacteroidetes; Bacteroidia; Bacteroidales; Rikenellaceae; Alistipes; Alistipes Alistipes senegalensis_msp_0381 0.11 0.16 0.02 0.11 Rikenellaceae senegalensis Bacteroidetes; Bacteroidia; Bacteroidales; Tannerellaceae; Parabacteroides; Parabacteroides goldsteinii_msp_0028 0.2 0.06 0.14 0.17 Tannerellaceae Parabacteroides goldsteinii Bacteroidetes; Bacteroidia; Bacteroidales; Tannerellaceae; Parabacteroides; Parabacteroides merdae_msp_0027 -0.02 -0.15 -0.05 -0.09 Tannerellaceae Parabacteroides merdae Firmicutes; Bacilli; Lactobacillales; Streptococcaceae; Streptococcus; Streptococcus thermophilus_msp_0833 -0.16 -0.06 -0.23 -0.16 Streptococcaceae Streptococcus thermophilus Butyricicoccus sp. 2789STDY5834927 / Clostridia Firmicutes; Clostridia; Clostridiales; Clostridiaceae; Butyricicoccus; -0.06 -0.17 -0.03 -0.05 Clostridiaceae bacterium UC5.1-1D1_msp_1381 Butyricicoccus sp. 2789STDY5834927 / Clostridia bacterium UC5.1-1D1 Firmicutes; Clostridia; Clostridiales; Clostridiaceae; Clostridium; Clostridium Clostridium saudiense_msp_0362 -0.15 -0.17 -0.07 -0.17 Clostridiaceae saudiense Firmicutes; Clostridia; Clostridiales; Clostridiaceae; Clostridium; Clostridium Clostridium sp. 2789STDY5608831_msp_1608c -0.11 0.16 -0.03 -0.02 Clostridiaceae sp. 2789STDY5608831 Firmicutes; Clostridia; Clostridiales; Clostridiaceae; Clostridium; Clostridium Clostridium sp. AT4_msp_0066 -0.14 -0.14 -0.14 -0.17 Clostridiaceae sp. AT4 Firmicutes; Clostridia; Clostridiales; Clostridiaceae; Clostridium; unclassified unclassified Clostridium_msp_0563 0.19 -0.02 0.12 0.13 Clostridiaceae Clostridium Firmicutes; Clostridia; Clostridiales; Eubacteriaceae; Eubacterium; Eubacterium Eubacterium eligens_msp_0029 0.18 0.2 0.25 0.26 Eubacteriaceae eligens Meslier V, et al. Gut 2020; 69:1258–1268. doi: 10.1136/gutjnl-2019-320438 Supplementary material Gut Firmicutes; Clostridia; Clostridiales; Eubacteriaceae; Eubacterium; Eubacterium Eubacterium sp. 36_13 & CAG:86_msp_0227 0.19 0.14 0.24 0.24 Eubacteriaceae sp. 36_13 & CAG:86 Firmicutes; Clostridia; Clostridiales; Eubacteriaceae; Eubacterium; Eubacterium Eubacterium sp. CAG:115_msp_0047 0.21 0.06 0.16 0.22 Eubacteriaceae sp. CAG:115 Firmicutes; Clostridia; Clostridiales; Eubacteriaceae; Eubacterium; Eubacterium Eubacterium sp. CAG:180_msp_0271 -0.19 -0.05 -0.13 -0.18 Eubacteriaceae sp. CAG:180 Firmicutes; Clostridia; Clostridiales; Eubacteriaceae; Eubacterium; Eubacterium Eubacterium sp. CAG:248_msp_0161 -0.02 -0.01 -0.15 -0.05 Eubacteriaceae sp. CAG:248 Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; Blautia Blautia massiliensis_msp_0141 -0.18 -0.2 -0.13 -0.21 Lachnospiraceae massiliensis Blautia obeum_msp_0436 0.02 0.15 0.04 0.06 Lachnospiraceae Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; Blautia obeum Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; Blautia sp. Blautia sp. CAG:237_msp_0189 0.21 -0.03 0.19 0.19 Lachnospiraceae CAG:237 Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; Blautia sp. Blautia sp. CAG:257_msp_0052 -0.19 -0.15 -0.15 -0.24 Lachnospiraceae CAG:257 Blautia sp. Marseille-P3087 / Ruminococcus sp. Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; Blautia sp. -0.16 -0.08 -0.01 -0.14 Lachnospiraceae 2789STDY5608882_msp_0722 Marseille-P3087 / Ruminococcus sp. 2789STDY5608882 Blautia wexlerae_msp_0076 -0.17 -0.25 -0.1 -0.21 Lachnospiraceae Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; Blautia wexlerae Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; Ruminococcus Ruminococcus gnavus_msp_0058 -0.2 -0.21 -0.24 -0.26 Lachnospiraceae gnavus Ruminococcus sp. CAG:60 / Blautia sp. Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; Ruminococcus 0.23 0.09 0.15 0.19 Lachnospiraceae 2789STDY5608836_msp_0244 sp. CAG:60 / Blautia sp. 2789STDY5608836 Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; Ruminococcus Ruminococcus torques_msp_0126 -0.13 -0.17 -0.15 -0.15 Lachnospiraceae torques Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; unclassified unclassified Blautia_msp_0756 0.14 0.23 0.15 0.16 Lachnospiraceae Blautia Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Blautia; unclassified unclassified Blautia_msp_1385 0 0.15 -0.04 0.04 Lachnospiraceae Blautia Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Coprococcus; Coprococcus catus_msp_0986 -0.01 0.01 0.19 0 Lachnospiraceae Coprococcus catus Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Coprococcus; Coprococcus comes_msp_0259 -0.2 -0.03 -0.12 -0.15 Lachnospiraceae Coprococcus comes Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Coprococcus; Coprococcus eutactus 2_msp_0042 0.13 0.16 0.03 0.14 Lachnospiraceae Coprococcus eutactus 2 Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Dorea; Dorea Dorea formicigenerans_msp_0506 -0.22 -0.15 -0.04 -0.22 Lachnospiraceae formicigenerans Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Dorea; Dorea Dorea longicatena 1_msp_0307 -0.24 -0.03 -0.06 -0.19 Lachnospiraceae longicatena 1 Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Eisenbergiella; Eisenbergiella tayi_msp_0014 -0.2 -0.09 0 -0.16 Lachnospiraceae Eisenbergiella tayi Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Lachnoclostridium; Clostridium asparagiforme == lavalense_msp_0360 0.09 -0.24 0.04 0.04 Lachnospiraceae Clostridium asparagiforme == lavalense Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Lachnoclostridium; Clostridium bolteae_msp_0009 -0.04 -0.26 -0.02 -0.07 Lachnospiraceae Clostridium bolteae Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Lachnoclostridium; Clostridium clostridioforme 1_msp_0020 -0.09 -0.09 -0.15 -0.08 Lachnospiraceae Clostridium clostridioforme 1 Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Lachnoclostridium; Clostridium glycyrrhizinilyticum_msp_0510 -0.16 -0.14 -0.12 -0.2 Lachnospiraceae Clostridium glycyrrhizinilyticum Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Lachnoclostridium; Clostridium symbiosum_msp_0086 -0.01 -0.17 -0.08 -0.05 Lachnospiraceae Clostridium symbiosum Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Lachnoclostridium; unclassified Lachnoclostridium_msp_0049 0.22 -0.06 0.03 0.18 Lachnospiraceae unclassified Lachnoclostridium Meslier V, et al. Gut 2020; 69:1258–1268. doi: 10.1136/gutjnl-2019-320438 Supplementary material Gut Lachnospira sp. 2789STDY5834967 / Clostridiales Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Lachnospira; bacterium KLE1615 & 0.15 0.09 0.23 0.19 Lachnospiraceae Lachnospira sp. 2789STDY5834967 / Clostridiales bacterium KLE1615 & 41_12_two_minus_msp_0572 41_12_two_minus Firmicutes; Clostridia; Clostridiales; Lachnospiraceae; Roseburia; Roseburia Roseburia faecis_msp_0015 0.15 -0.03 0.11 0.17 Lachnospiraceae faecis Firmicutes; Clostridia; Clostridiales;
Recommended publications
  • Official Nh Dhhs Health Alert
    THIS IS AN OFFICIAL NH DHHS HEALTH ALERT Distributed by the NH Health Alert Network [email protected] May 18, 2018, 1300 EDT (1:00 PM EDT) NH-HAN 20180518 Tickborne Diseases in New Hampshire Key Points and Recommendations: 1. Blacklegged ticks transmit at least five different infections in New Hampshire (NH): Lyme disease, Anaplasma, Babesia, Powassan virus, and Borrelia miyamotoi. 2. NH has one of the highest rates of Lyme disease in the nation, and 50-60% of blacklegged ticks sampled from across NH have been found to be infected with Borrelia burgdorferi, the bacterium that causes Lyme disease. 3. NH has experienced a significant increase in human cases of anaplasmosis, with cases more than doubling from 2016 to 2017. The reason for the increase is unknown at this time. 4. The number of new cases of babesiosis also increased in 2017; because Babesia can be transmitted through blood transfusions in addition to tick bites, providers should ask patients with suspected babesiosis whether they have donated blood or received a blood transfusion. 5. Powassan is a newer tickborne disease which has been identified in three NH residents during past seasons in 2013, 2016 and 2017. While uncommon, Powassan can cause a debilitating neurological illness, so providers should maintain an index of suspicion for patients presenting with an unexplained meningoencephalitis. 6. Borrelia miyamotoi infection usually presents with a nonspecific febrile illness similar to other tickborne diseases like anaplasmosis, and has recently been identified in one NH resident. Tests for Lyme disease do not reliably detect Borrelia miyamotoi, so providers should consider specific testing for Borrelia miyamotoi (see Attachment 1) and other pathogens if testing for Lyme disease is negative but a tickborne disease is still suspected.
    [Show full text]
  • The Influence of Probiotics on the Firmicutes/Bacteroidetes Ratio In
    microorganisms Review The Influence of Probiotics on the Firmicutes/Bacteroidetes Ratio in the Treatment of Obesity and Inflammatory Bowel disease Spase Stojanov 1,2, Aleš Berlec 1,2 and Borut Štrukelj 1,2,* 1 Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia; [email protected] (S.S.); [email protected] (A.B.) 2 Department of Biotechnology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia * Correspondence: borut.strukelj@ffa.uni-lj.si Received: 16 September 2020; Accepted: 31 October 2020; Published: 1 November 2020 Abstract: The two most important bacterial phyla in the gastrointestinal tract, Firmicutes and Bacteroidetes, have gained much attention in recent years. The Firmicutes/Bacteroidetes (F/B) ratio is widely accepted to have an important influence in maintaining normal intestinal homeostasis. Increased or decreased F/B ratio is regarded as dysbiosis, whereby the former is usually observed with obesity, and the latter with inflammatory bowel disease (IBD). Probiotics as live microorganisms can confer health benefits to the host when administered in adequate amounts. There is considerable evidence of their nutritional and immunosuppressive properties including reports that elucidate the association of probiotics with the F/B ratio, obesity, and IBD. Orally administered probiotics can contribute to the restoration of dysbiotic microbiota and to the prevention of obesity or IBD. However, as the effects of different probiotics on the F/B ratio differ, selecting the appropriate species or mixture is crucial. The most commonly tested probiotics for modifying the F/B ratio and treating obesity and IBD are from the genus Lactobacillus. In this paper, we review the effects of probiotics on the F/B ratio that lead to weight loss or immunosuppression.
    [Show full text]
  • The 2014 Golden Gate National Parks Bioblitz - Data Management and the Event Species List Achieving a Quality Dataset from a Large Scale Event
    National Park Service U.S. Department of the Interior Natural Resource Stewardship and Science The 2014 Golden Gate National Parks BioBlitz - Data Management and the Event Species List Achieving a Quality Dataset from a Large Scale Event Natural Resource Report NPS/GOGA/NRR—2016/1147 ON THIS PAGE Photograph of BioBlitz participants conducting data entry into iNaturalist. Photograph courtesy of the National Park Service. ON THE COVER Photograph of BioBlitz participants collecting aquatic species data in the Presidio of San Francisco. Photograph courtesy of National Park Service. The 2014 Golden Gate National Parks BioBlitz - Data Management and the Event Species List Achieving a Quality Dataset from a Large Scale Event Natural Resource Report NPS/GOGA/NRR—2016/1147 Elizabeth Edson1, Michelle O’Herron1, Alison Forrestel2, Daniel George3 1Golden Gate Parks Conservancy Building 201 Fort Mason San Francisco, CA 94129 2National Park Service. Golden Gate National Recreation Area Fort Cronkhite, Bldg. 1061 Sausalito, CA 94965 3National Park Service. San Francisco Bay Area Network Inventory & Monitoring Program Manager Fort Cronkhite, Bldg. 1063 Sausalito, CA 94965 March 2016 U.S. Department of the Interior National Park Service Natural Resource Stewardship and Science Fort Collins, Colorado The National Park Service, Natural Resource Stewardship and Science office in Fort Collins, Colorado, publishes a range of reports that address natural resource topics. These reports are of interest and applicability to a broad audience in the National Park Service and others in natural resource management, including scientists, conservation and environmental constituencies, and the public. The Natural Resource Report Series is used to disseminate comprehensive information and analysis about natural resources and related topics concerning lands managed by the National Park Service.
    [Show full text]
  • Naeglaria and Brain Infections
    Can bacteria shrink tumors? Cancer Therapy: The Microbial Approach n this age of advanced injected live Streptococcus medical science and into cancer patients but after I technology, we still the recipients unfortunately continue to hunt for died from subsequent innovative cancer therapies infections, Coley decided to that prove effective and safe. use heat killed bacteria. He Treatments that successfully made a mixture of two heat- eradicate tumors while at the killed bacterial species, By Alan Barajas same time cause as little Streptococcus pyogenes and damage as possible to normal Serratia marcescens. This Alani Barajas is a Research and tissue are the ultimate goal, concoction was termed Development Technician at Hardy but are also not easy to find. “Coley’s toxins.” Bacteria Diagnostics. She earned her bachelor's degree in Microbiology at were either injected into Cal Poly, San Luis Obispo. The use of microorganisms in tumors or into the cancer therapy is not a new bloodstream. During her studies at Cal Poly, much idea but it is currently a of her time was spent as part of the undergraduate research team for the buzzing topic in cancer Cal Poly Dairy Products Technology therapy research. Center studying spore-forming bacteria in dairy products. In the late 1800s, German Currently she is working on new physicians W. Busch and F. chromogenic media formulations for Fehleisen both individually Hardy Diagnostics, both in the observed that certain cancers prepared and powdered forms. began to regress when patients acquired accidental erysipelas (cellulitis) caused by Streptococcus pyogenes. William Coley was the first to use New York surgeon William bacterial injections to treat cancer www.HardyDiagnostics.com patients.
    [Show full text]
  • Fatty Acid Diets: Regulation of Gut Microbiota Composition and Obesity and Its Related Metabolic Dysbiosis
    International Journal of Molecular Sciences Review Fatty Acid Diets: Regulation of Gut Microbiota Composition and Obesity and Its Related Metabolic Dysbiosis David Johane Machate 1, Priscila Silva Figueiredo 2 , Gabriela Marcelino 2 , Rita de Cássia Avellaneda Guimarães 2,*, Priscila Aiko Hiane 2 , Danielle Bogo 2, Verônica Assalin Zorgetto Pinheiro 2, Lincoln Carlos Silva de Oliveira 3 and Arnildo Pott 1 1 Graduate Program in Biotechnology and Biodiversity in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; [email protected] (D.J.M.); [email protected] (A.P.) 2 Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; pri.fi[email protected] (P.S.F.); [email protected] (G.M.); [email protected] (P.A.H.); [email protected] (D.B.); [email protected] (V.A.Z.P.) 3 Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; [email protected] * Correspondence: [email protected]; Tel.: +55-67-3345-7416 Received: 9 March 2020; Accepted: 27 March 2020; Published: 8 June 2020 Abstract: Long-term high-fat dietary intake plays a crucial role in the composition of gut microbiota in animal models and human subjects, which affect directly short-chain fatty acid (SCFA) production and host health. This review aims to highlight the interplay of fatty acid (FA) intake and gut microbiota composition and its interaction with hosts in health promotion and obesity prevention and its related metabolic dysbiosis.
    [Show full text]
  • Gut Microbiota Differences Between Psoriatic Arthritis and Undifferentiated Arthritis Patients
    Gut microbiota differences between psoriatic arthritis and undifferentiated arthritis patients Chung-Yuan Hsu Chang Gung Memorial Hospital Kaohsiung Branch Ho-Chang Kuo Chang Gung Memorial Hospital Kaohsiung Branch YU-JIH Su ( [email protected] ) Chang Gung Memorial Hospital Kaohsiung Branch https://orcid.org/0000-0002-7274-3458 Research article Keywords: Psoriatic arthritis, undifferentiated arthritis, enthesitis, dactylitis, microbiota Posted Date: February 28th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-15400/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/15 Abstract Introduction Psoriatic arthritis (PSA) is a form of immune-mediated inammatory arthritis. Studying the gut microbiota of PSA patients may offer new insights into the pathophysiology of this inammatory joint disease. We designed a prospective study to examine gut microbiome from patients with PSA, primarily with enthesitis and dactylitis, and compared the data with undifferentiated arthritis patients (NO PSA), without enthesitis or dactylitis. Methods We enrolled nine PSA patients and 10 NO PSA patients in this study. The fecal samples were investigated by using 16S rRNA amplicon sequencing, followed by bioinformatics and statistical analyses. Results None of the available objective clinical laboratory data could differentiate PSA from the NO PSA subgroup. The microbiota result shows that Family: XIII_AD3011 is signicantly higher in NO PSA patients than PSA patients’ stool samples (p=0.039). Megasphaera elsdenii in the PSA was 10000 times higher than in the NO PSA group. Conclusion Our results demonstrated high intra-group homogeneous and high inter-group heterogeneous microbiota. The clinical symptoms of either enthesitis or dactylitis link to the specic microbiota in the current study.
    [Show full text]
  • Appendix III: OTU's Found to Be Differentially Abundant Between CD and Control Patients Via Metagenomeseq Analysis
    Appendix III: OTU's found to be differentially abundant between CD and control patients via metagenomeSeq analysis OTU Log2 (FC CD / FDR Adjusted Phylum Class Order Family Genus Species Number Control) p value 518372 Firmicutes Clostridia Clostridiales Ruminococcaceae Faecalibacterium prausnitzii 2.16 5.69E-08 194497 Firmicutes Clostridia Clostridiales Ruminococcaceae NA NA 2.15 8.93E-06 175761 Firmicutes Clostridia Clostridiales Ruminococcaceae NA NA 5.74 1.99E-05 193709 Firmicutes Clostridia Clostridiales Ruminococcaceae NA NA 2.40 2.14E-05 4464079 Bacteroidetes Bacteroidia Bacteroidales Bacteroidaceae Bacteroides NA 7.79 0.000123188 20421 Firmicutes Clostridia Clostridiales Lachnospiraceae Coprococcus NA 1.19 0.00013719 3044876 Firmicutes Clostridia Clostridiales Lachnospiraceae [Ruminococcus] gnavus -4.32 0.000194983 184000 Firmicutes Clostridia Clostridiales Ruminococcaceae Faecalibacterium prausnitzii 2.81 0.000306032 4392484 Bacteroidetes Bacteroidia Bacteroidales Bacteroidaceae Bacteroides NA 5.53 0.000339948 528715 Firmicutes Clostridia Clostridiales Ruminococcaceae Faecalibacterium prausnitzii 2.17 0.000722263 186707 Firmicutes Clostridia Clostridiales NA NA NA 2.28 0.001028539 193101 Firmicutes Clostridia Clostridiales Ruminococcaceae NA NA 1.90 0.001230738 339685 Firmicutes Clostridia Clostridiales Peptococcaceae Peptococcus NA 3.52 0.001382447 101237 Firmicutes Clostridia Clostridiales NA NA NA 2.64 0.001415109 347690 Firmicutes Clostridia Clostridiales Ruminococcaceae Oscillospira NA 3.18 0.00152075 2110315 Firmicutes Clostridia
    [Show full text]
  • Effect of Ph and Temperature on Microbial Community Structure And
    Calicioglu et al. Biotechnol Biofuels (2018) 11:275 https://doi.org/10.1186/s13068-018-1278-6 Biotechnology for Biofuels RESEARCH Open Access Efect of pH and temperature on microbial community structure and carboxylic acid yield during the acidogenic digestion of duckweed Ozgul Calicioglu1, Michael J. Shreve1, Tom L. Richard2 and Rachel A. Brennan1* Abstract Background: Duckweeds (Lemnaceae) are efcient aquatic plants for wastewater treatment due to their high nutri- ent-uptake capabilities and resilience to severe environmental conditions. Combined with their rapid growth rates, high starch, and low lignin contents, duckweeds have also gained popularity as a biofuel feedstock for thermochemi- cal conversion and alcohol fermentation. However, studies on the acidogenic anaerobic digestion of duckweed into carboxylic acids, another group of chemicals which are precursors of higher-value chemicals and biofuels, are lacking. In this study, a series of laboratory batch experiments were performed to determine the favorable operating condi- tions (i.e., temperature and pH) to maximize carboxylic acid production from wastewater-derived duckweed during acidogenic digestion. Batch reactors with 25 g/l solid loading were operated anaerobically for 21 days under meso- philic (35 °C) or thermophilic (55 °C) conditions at an acidic (5.3) or basic (9.2) pH. At the conclusion of the experiment, the dominant microbial communities under various operating conditions were assessed using high-throughput sequencing. Results: The highest duckweed–carboxylic acid conversion of 388 28 mg acetic acid equivalent per gram volatile solids was observed under mesophilic and basic conditions, with an± average production rate of 0.59 g/l/day. This result is comparable to those reported for acidogenic digestion of other organics such as food waste.
    [Show full text]
  • Gut Microbiota and Inflammation
    Nutrients 2011, 3, 637-682; doi:10.3390/nu3060637 OPEN ACCESS nutrients ISSN 2072-6643 www.mdpi.com/journal/nutrients Review Gut Microbiota and Inflammation Asa Hakansson and Goran Molin * Food Hygiene, Division of Applied Nutrition, Department of Food Technology, Engineering and Nutrition, Lund University, PO Box 124, SE-22100 Lund, Sweden; E-Mail: [email protected] * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +46-46-222-8327; Fax: +46-46-222-4532. Received: 15 April 2011; in revised form: 19 May 2011 / Accepted: 24 May 2011 / Published: 3 June 2011 Abstract: Systemic and local inflammation in relation to the resident microbiota of the human gastro-intestinal (GI) tract and administration of probiotics are the main themes of the present review. The dominating taxa of the human GI tract and their potential for aggravating or suppressing inflammation are described. The review focuses on human trials with probiotics and does not include in vitro studies and animal experimental models. The applications of probiotics considered are systemic immune-modulation, the metabolic syndrome, liver injury, inflammatory bowel disease, colorectal cancer and radiation-induced enteritis. When the major genomic differences between different types of probiotics are taken into account, it is to be expected that the human body can respond differently to the different species and strains of probiotics. This fact is often neglected in discussions of the outcome of clinical trials with probiotics. Keywords: probiotics; inflammation; gut microbiota 1. Inflammation Inflammation is a defence reaction of the body against injury. The word inflammation originates from the Latin word ―inflammatio‖ which means fire, and traditionally inflammation is characterised by redness, swelling, pain, heat and impaired body functions.
    [Show full text]
  • A Metagenomic Study of the Oral and Gut Microbiome in Crohn’S Disease Shijia Hu1* , Eileen Png2, Michelle Gowans3, David E
    Hu et al. Gut Pathog (2021) 13:13 https://doi.org/10.1186/s13099-021-00409-5 Gut Pathogens RESEARCH Open Access Ectopic gut colonization: a metagenomic study of the oral and gut microbiome in Crohn’s disease Shijia Hu1* , Eileen Png2, Michelle Gowans3, David E. H. Ong3, Paola Florez de Sessions2, Jie Song2 and Niranjan Nagarajan2,4 Abstract Background: This study aims to characterize, the gut and oral microbiome in Asian subjects with Crohn’s disease (CD) using whole genome shotgun sequencing, thereby allowing for strain-level comparison. Methods: A case–control study with age, sex and ethnicity matched healthy controls was conducted. CD subjects were limited to well-controlled patients without oral manifestations. Fecal and saliva samples were collected for char- acterization of gut and oral microbiome respectively. Microbial DNA were extracted, libraries prepared and sequenced reads profled. Taxonomic diversity, taxonomic association, strain typing and microbial gene pathway analyses were conducted. Results: The study recruited 25 subjects with CD and 25 healthy controls. The oral microbe Streptococcus salivarius was found to be enriched and of concordant strains in the gut and oral microbiome of Crohn’s disease subjects. This was more likely in CD subjects with higher Crohn’s Disease Activity Index (184.3 2.9 vs 67.1 82.5, p 0.012) and active disease status (Diarrhoea/abdominal pain/blood-in-stool/fever and fatigue)± (p 0.016).± Gut species= found to be signifcantly depleted in CD compared to control (Relative abundance: Median[Range])= include: Faecalibacterium prausnitzii (0.03[0.00–4.56] vs 13.69[5.32–18.71], p 0.010), Roseburia inulinivorans (0.00[0.00–0.03] vs 0.21[0.01–0.53], p 0.010) and Alistipes senegalensis (0.00[0.00–0.00]= vs 0.00[0.00–0.02], p 0.029).
    [Show full text]
  • Gut Microbiota Features Associated with Clostridioides Difficile Colonization in Dairy Calves
    bioRxiv preprint doi: https://doi.org/10.1101/2021.05.11.443551; this version posted May 11, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 Gut microbiota features associated with Clostridioides 2 difficile colonization in dairy calves 3 4 Laurel E. Redding1, Alexander S. Berry2,3, Nagaraju Indugu1, Elizabeth Huang1, Daniel P. Beiting3, Dipti 5 Pitta1 6 7 8 1Department of Clinical Sciences, University of Pennsylvania, School of Veterinary Medicine, Kennett 9 Square, PA, USA 10 11 2Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, 12 PA, USA 13 14 3Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 15 USA 16 17 18 Corresponding author: Laurel Redding, [email protected] 19 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.05.11.443551; this version posted May 11, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 20 Abstract 21 Diarrheal disease, a major cause of morbidity and mortality in dairy calves, is strongly associated with the 22 health and composition of the gut microbiome. Clostridioides difficile is an opportunistic pathogen that 23 proliferates and can produce enterotoxins when the host experiences gut dysbiosis.
    [Show full text]
  • The Isolation of Novel Lachnospiraceae Strains and the Evaluation of Their Potential Roles in Colonization Resistance Against Clostridium Difficile
    The isolation of novel Lachnospiraceae strains and the evaluation of their potential roles in colonization resistance against Clostridium difficile Diane Yuan Wang Honors Thesis in Biology Department of Ecology and Evolutionary Biology College of Literature, Science, & the Arts University of Michigan, Ann Arbor April 1st, 2014 Sponsor: Vincent B. Young, M.D., Ph.D. Associate Professor of Internal Medicine Associate Professor of Microbiology and Immunology Medical School Co-Sponsor: Aaron A. King, Ph.D. Associate Professor of Ecology & Evolutionary Associate Professor of Mathematics College of Literature, Science, & the Arts Reader: Blaise R. Boles, Ph.D. Assistant Professor of Molecular, Cellular and Developmental Biology College of Literature, Science, & the Arts 1 Table of Contents Abstract 3 Introduction 4 Clostridium difficile 4 Colonization Resistance 5 Lachnospiraceae 6 Objectives 7 Materials & Methods 9 Sample Collection 9 Bacterial Isolation and Selective Growth Conditions 9 Design of Lachnospiraceae 16S rRNA-encoding gene primers 9 DNA extraction and 16S ribosomal rRNA-encoding gene sequencing 10 Phylogenetic analyses 11 Direct inhibition 11 Bile salt hydrolase (BSH) detection 12 PCR assay for bile acid 7α-dehydroxylase detection 12 Tables & Figures Table 1 13 Table 2 15 Table 3 21 Table 4 25 Figure 1 16 Figure 2 19 Figure 3 20 Figure 4 24 Figure 5 26 Results 14 Isolation of novel Lachnospiraceae strains 14 Direct inhibition 17 Bile acid physiology 22 Discussion 27 Acknowledgments 33 References 34 2 Abstract Background: Antibiotic disruption of the gastrointestinal tract’s indigenous microbiota can lead to one of the most common nosocomial infections, Clostridium difficile, which has an annual cost exceeding $4.8 billion dollars.
    [Show full text]