1
|
Bratseth V, Nendl A, Raju SC, Holm K, Broch K, Hov JR, Seljeflot I, Trøseid M, Awoyemi A. Gut dysbiosis and neutrophil extracellular traps in chronic heart failure. Int J Cardiol 2025; 419:132689. [PMID: 39489348 DOI: 10.1016/j.ijcard.2024.132689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/15/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Chronic heart failure (HF) patients have reduced microbiota diversity. Leakage of microbes and their metabolites into the bloodstream may activate neutrophils. Neutrophil extracellular traps (NETs) consist of chromatin and proteases, and may contribute to HF pathogenesis. We assessed associations between circulating NETs and 1) cardiac function, 2) the degree of gut microbiota diversity and 3) gut leakage and microbial metabolites in HF patients. METHODS A cross-sectional study including 124 patients with chronic HF and left ventricular ejection fraction ≤40 %. Severe HF was defined as N-terminal pro-B-type natriuretic peptide concentrations above median. We measured citrullinated histone H3 (CitH3), myeloperoxidase- and double-stranded-DNA in the blood. Gut leakage markers included bacterial lipopolysaccharides and soluble cluster of differentiation 14. The microbial metabolites included circulating trimethylamine N-oxide and butyrate producing capacity. We used the Shannon diversity-index and a dysbiosis-index based on bacteria with altered relative abundance to characterize the gut microbiota profile. RESULTS Quartile 4 of CitH3 was associated with more severe HF compared to quartiles 1-3, after adjustments for age, gender and hypertension (adjusted odds ratio [95 %CI] 3.21[1.18-8.69], p = 0.022). CitH3 was moderately associated with hypertension (p = 0.04), higher CRP levels (p = 0.016) and lower Shannon diversity index, (p = 0.039). No other NET marker associated with severe HF. CONCLUSIONS In chronic HF patients with reduced LVEF, high levels of CitH3 were associated with disease severity, inflammation and reduced gut microbiota diversity. Our results suggest that enhanced release of NETs could be involved in progressive HF, although the contribution of the gut microbiota seems limited in this context.
Collapse
Affiliation(s)
- Vibeke Bratseth
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, Oslo, Norway.
| | - Andraz Nendl
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sajan C Raju
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kristian Holm
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway; Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Johannes R Hov
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway; Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway; Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Ingebjørg Seljeflot
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marius Trøseid
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Section of Clinical Immunology, and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ayodeji Awoyemi
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, Oslo, Norway; Department of Cardiology Ullevaal, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
2
|
Menon R, Bhattarai SK, Crossette E, Prince AL, Olle B, Silber JL, Bucci V, Faith J, Norman JM. Multi-omic profiling a defined bacterial consortium for treatment of recurrent Clostridioides difficile infection. Nat Med 2025:10.1038/s41591-024-03337-4. [PMID: 39747680 DOI: 10.1038/s41591-024-03337-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 10/02/2024] [Indexed: 01/04/2025]
Abstract
Donor-derived fecal micrrasobiota treatments are efficacious in preventing recurrent Clostridioides difficile infection (rCDI), but they have inherently variable quality attributes, are difficult to scale and harbor the risk of pathogen transfer. In contrast, VE303 is a defined consortium of eight purified, clonal bacterial strains developed for prevention of rCDI. In the phase 2 CONSORTIUM study, high-dose VE303 was well tolerated and reduced the odds of rCDI by more than 80% compared to placebo. VE303 organisms robustly colonized the gut in the high-dose group and were among the top taxa associated with non-recurrence. Multi-omic modeling identified antibiotic history, baseline stool metabolites and serum cytokines as predictors of both on-study CDI recurrence and VE303 colonization. VE303 potentiated early recovery of the host microbiome and metabolites with increases in short-chain fatty acids, secondary bile acids and bile salt hydrolase genes after antibiotic treatment for CDI, which is considered important to prevent CDI recurrences. These results support the idea that VE303 promotes efficacy in rCDI through multiple mechanisms.
Collapse
Affiliation(s)
| | - Shakti K Bhattarai
- Program in Microbiome Dynamics, Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | | | - Bernat Olle
- Vedanta Biosciences, Inc., Cambridge, MA, USA
| | | | - Vanni Bucci
- Program in Microbiome Dynamics, Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeremiah Faith
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
3
|
Yang J, Lei OK, Bhute S, Kris-Etherton PM, Lichtenstein AH, Matthan NR, Petersen KS, Sabaté J, Reboussin DM, Lovato L, Vitolins MZ, Rajaram S, Jacobs JP, Huang J, Taw M, Yang S, Li Z. Impact of daily avocado consumption on gut microbiota in adults with abdominal obesity: an ancillary study of HAT, a randomized controlled trial. Food Funct 2025; 16:168-180. [PMID: 39641169 DOI: 10.1039/d4fo03806a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Objectives: This study aimed to investigate short-term and long-term impact of avocado consumption without caloric restriction on the gut microbiota of free-living adults with abdominal obesity. Methods: The Habitual Diet and Avocado Trial (HAT) was a 26-week, multi-center, randomized, controlled trial involving 1008 individuals with abdominal obesity. Participants were randomly assigned to the Avocado Supplemented Diet Group (AVO), receiving one avocado per day, or the Habitual Diet group (HAB), maintaining their usual dietary habits. Fecal samples were collected at baseline, week 4 and week 26 from a subset of participants recruited at a University of California Los Angeles site (n = 230). Fecal microbiota was assessed with shotgun metagenomics sequencing. Alpha diversity was assessed using the Chao1 and Shannon indices; beta diversity was assessed using Bray-Curtis dissimilarity with significance determined by repeated measures permutational multivariat analysis of variance. Potential association of intervention at week 4 and 26 with alpha diversity, species and metabolic pathways was examined using linear mixed effect models. Results: Compared to the HAB group, the AVO group had higher alpha diversity by 4 weeks, which persisted through the 26-week study period. Exploratory analysis based on healthy eating index-2015 (HEI-2015) indicated that participants with a low HEI score at baseline (≤52.7), had an increase in alpha diversity in the AVO group vs. HAB group. The AVO group had a significant change in beta diversity at week 26 compared to the HAB group. At the species level, the AVO group had significantly increased Faecalibacterium prausnitzii and Bacterium AF16_15 at week 26 compared to the HAB group. Functional analysis showed no significant difference in metabolic pathways between the HAB and AVO groups. Conclusions: Our findings document a potentially favorable effect of avocados on gut microbiota diversity. The prebiotic potential of avocados is more pronounced in individuals with a low diet quality score. This trial is registered at clinicaltrials.gov as NCT03528031 (https://clinicaltrials.gov/study/NCT03528031).
Collapse
Affiliation(s)
- Jieping Yang
- Center for Human Nutrition, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA, 90095, USA.
| | - On Kei Lei
- Center for Human Nutrition, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA, 90095, USA.
- Faculty of Education, University of Macau, Macao, China
| | - Shrikant Bhute
- David Geffen School of Medicine, Department of Medicine, Los Angeles, CA, 90095, USA
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Nirupa R Matthan
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Kristina S Petersen
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Joan Sabaté
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, CA, 92354, USA
| | - David M Reboussin
- Department of Biostatistics, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Laura Lovato
- Department of Biostatistics, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Mara Z Vitolins
- Department of Biostatistics, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Sujatha Rajaram
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, CA, 92354, USA
| | - Jonathan P Jacobs
- David Geffen School of Medicine, Department of Medicine, Los Angeles, CA, 90095, USA
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Medicine VA Greater Los Angeles HealthCare System, Los Angeles, CA 90073, USA
| | - Jianjun Huang
- Center for Human Nutrition, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA, 90095, USA.
| | - Meileen Taw
- Center for Human Nutrition, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA, 90095, USA.
| | - Scarlet Yang
- Center for Human Nutrition, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA, 90095, USA.
| | - Zhaoping Li
- Center for Human Nutrition, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA, 90095, USA.
- Department of Medicine VA Greater Los Angeles HealthCare System, Los Angeles, CA 90073, USA
| |
Collapse
|
4
|
Aydin Ö, Wahlström A, de Jonge PA, Meijnikman AS, Sjöland W, Olsson L, Henricsson M, de Goffau MC, Oonk S, Bruin SC, Acherman YIZ, Marschall HU, Gerdes VEA, Nieuwdorp M, Bäckhed F, Groen AK. An integrated analysis of bile acid metabolism in humans with severe obesity. Hepatology 2025; 81:19-31. [PMID: 39010331 DOI: 10.1097/hep.0000000000000938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 03/26/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND AND AIMS Bile acids (BA) are vital regulators of metabolism. BAs are AQ6 secreted in the small intestine, reabsorbed, and transported back to the liver, where they can modulate metabolic functions. There is a paucity of data regarding the portal BA composition in humans. This study aimed to address this knowledge gap by investigating portal BA composition and the relation with peripheral and fecal BA dynamics in conjunction with the gut microbiome. APPROACH AND RESULTS Thirty-three individuals from the BARIA cohort were included. Portal plasma, peripheral plasma, and feces were collected. BA and C4 levels were measured employing mass spectrometry. FGF19 was measured using ELISA. Gut microbiota composition was determined through metagenomics analysis on stool samples. Considerable diversity in the portal BA composition was observed. The majority (n = 26) of individuals had a 9-fold higher portal than peripheral BA concentration. In contrast, 8 individuals showed lower portal BA concentration compared with peripheral and had higher levels of unconjugated and secondary BA in this compartment, suggesting more distal origin. The altered portal BA profile was associated with altered gut microbiota composition. In particular, taxa within Bacteroides were reduced in abundance in the feces of these individuals. CONCLUSIONS Characterization of the portal BA composition in relation to peripheral and fecal BA increased insight into the dynamics of BA metabolism in individuals with obesity. Peripheral BA composition was much more diverse due to microbial metabolism. About 24% of the portal samples was surprisingly low in total BA; the underlying mechanism requires further exploration.
Collapse
Affiliation(s)
- Ömrüm Aydin
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Annika Wahlström
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Patrick A de Jonge
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Abraham S Meijnikman
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Wilhelm Sjöland
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lisa Olsson
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Henricsson
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marcus C de Goffau
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Stijn Oonk
- Department of Scientific Research, Data Science, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
| | - Sjoerd C Bruin
- Department of Bariatric Surgery, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
| | - Yair I Z Acherman
- Department of Bariatric Surgery, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
| | - Hanns-Ulrich Marschall
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Victor E A Gerdes
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Bariatric Surgery, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
- Department of Internal Medicine, Spaarne Gasthuis Hospital, Hoofddorp, the Netherlands
| | - Max Nieuwdorp
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Fredrik Bäckhed
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Albert K Groen
- Department of Internal and (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, ACS Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Masson BA, Kiridena P, Lu D, Kleeman EA, Reisinger SN, Qin W, Davies WJ, Muralitharan RR, Jama HA, Antonacci S, Marques FZ, Gubert C, Hannan AJ. Depletion of the paternal gut microbiome alters sperm small RNAs and impacts offspring physiology and behavior in mice. Brain Behav Immun 2025; 123:290-305. [PMID: 39293692 DOI: 10.1016/j.bbi.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
The paternal environment prior to conception has been demonstrated to influence offspring physiology and behavior, with the sperm epigenome (including noncoding RNAs) proposed as a potential facilitator of non-genetic inheritance. Whilst the maternal gut microbiome has been established as an important influence on offspring development, the impact of the paternal gut microbiome on offspring development, health and behavior is largely unknown. Gut microbiota have major influences on immunity, and thus we hypothesized that they may be relevant to paternal immune activation (PIA) modulating epigenetic inheritance in mice. Therefore, male C57BL/6J mice (F0) were orally administered non-absorbable antibiotics via drinking water in order to substantially deplete their gut microbiome. Four weeks after administration of the antibiotics (gut microbiome depletion), F0 male mice were then mated with naïve female mice. The F1 offspring of the microbiome-depleted males had reduced body weight as well as altered gut morphology (shortened colon length). F1 females showed significant alterations in affective behaviors, including measures of anxiety and depressive-like behaviors, indicating altered development. Analysis of small noncoding RNAs in the sperm of F0 mice revealed that gut microbiome depletion is associated with differential expression of 8 different PIWI-interacting RNAs (piRNAs), each of which has the potential to modulate the expression of multiple downstream gene targets, and thus influence epigenetic inheritance and offspring development. This study demonstrates that the gut-germline axis influences sperm small RNA profiles and offspring physiology, with specific impacts on offspring affective and/or coping behaviors. These findings may have broader implications for other animal species with comparable gut microbiota, intergenerational epigenetics and developmental biology, including humans.
Collapse
Affiliation(s)
- Bethany A Masson
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Pamudika Kiridena
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Da Lu
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Sonali N Reisinger
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Wendy Qin
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - William J Davies
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Simona Antonacci
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
6
|
Olivera PA, Martinez-Lozano H, Leibovitzh H, Xue M, Neustaeter A, Espin-Garcia O, Xu W, Madsen KL, Guttman DS, Bernstein CN, Yerushalmi B, Hyams JS, Abreu MT, Marshall JK, Wrobel I, Mack DR, Jacobson K, Bitton A, Aumais G, Panacionne R, Dieleman LA, Silverberg MS, Steinhart AH, Moayyedi P, Turner D, Griffiths AM, Turpin W, Lee SH, Croitoru K. Healthy First-Degree Relatives From Multiplex Families vs Simplex Families Have Higher Subclinical Intestinal Inflammation, a Distinct Fecal Microbial Signature, and Harbor a Higher Risk of Developing Crohn's Disease. Gastroenterology 2025; 168:99-110.e2. [PMID: 39236898 DOI: 10.1053/j.gastro.2024.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 08/09/2024] [Accepted: 08/25/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND & AIMS Unaffected first-degree relatives (FDRs) from families with ≥2 affected FDRs with Crohn's disease (CD, multiplex families) have a high risk of developing CD, although the underlying mechanisms driving this risk are poorly understood. We aimed to identify differences in biomarkers between FDRs from multiplex vs simplex families and investigate the risk of future CD onset accounting for potential confounders. METHODS We assessed the Crohn's and Colitis Canada Genetic Environmental Microbial cohort of healthy FDRs of patients with CD. Genome-wide CD-polygenic risk scores, urinary fractional excretion of lactulose-to-mannitol ratio, fecal calprotectin (FCP), and fecal 16S ribosomal RNA microbiome were measured at recruitment. Associations between CD multiplex status and baseline biomarkers were determined using generalized estimating equations models. Cox models were used to assess the risk of future CD onset. RESULTS There were 4051 participants from simplex families and 334 from CD multiplex families. CD multiplex status was significantly associated with higher baseline FCP (P = .026) but not with baseline CD-polygenic risk scores or the lactulose-to-mannitol ratio. Three bacterial genera were found to be differentially abundant between both groups. CD multiplex status at recruitment was independently associated with an increased risk of developing CD (adjusted hazard ratio, 3.65; 95% confidence interval, 2.18-6.11, P < .001). CONCLUSION Within FDRs of patients with CD, participants from multiplex families had a 3-fold increased risk of CD onset, a higher FCP, and an altered bacterial composition, but not genetic burden or altered gut permeability. These results suggest that putative environmental factors might be enriched in FDRs from multiplex families.
Collapse
Affiliation(s)
- Pablo A Olivera
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, University of Toronto Temerty Faculty of Medicine, Toronto, Ontario, Canada
| | - Helena Martinez-Lozano
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, University of Toronto Temerty Faculty of Medicine, Toronto, Ontario, Canada; Department of Digestive System Medicine, Hospital General Universitario, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Haim Leibovitzh
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, University of Toronto Temerty Faculty of Medicine, Toronto, Ontario, Canada
| | - Mingyue Xue
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Anna Neustaeter
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Osvaldo Espin-Garcia
- Division of Biostatistics, University of Toronto Dalla Lana School of Public Health, Toronto, Ontario, Canada
| | - Wei Xu
- Division of Biostatistics, University of Toronto Dalla Lana School of Public Health, Toronto, Ontario, Canada
| | - Karen L Madsen
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - David S Guttman
- Department of Cell & Systems Biology and Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Charles N Bernstein
- Inflammatory Bowel Disease Clinical and Research Centre and Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Baruch Yerushalmi
- Pediatric Gastroenterology Unit, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Jeffrey S Hyams
- Division of Digestive Diseases, Hepatology, and Nutrition, Connecticut Children's Medical Center, Hartford, Connecticut
| | - Maria T Abreu
- Division of Gastroenterology, Crohn's and Colitis Center, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - John K Marshall
- Department of Medicine, Farncombe Family Digestive Health Research Institute McMaster University, Hamilton, Ontario, Canada
| | - Iwona Wrobel
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - David R Mack
- Division of Gastroenterology, Hepatology & Nutrition, Children's Hospital of Eastern Ontario and University of Ottawa, Ottawa, Ontario, Canada
| | - Kevan Jacobson
- Canadian Gastro-Intestinal Epidemiology Consortium, Toronto, Ontario, Canada; British Columbia Children's Hospital, Vancouver, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alain Bitton
- Division of Gastroenterology and Hepatology, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Guy Aumais
- Department of Medicine, Montreal University, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Remo Panacionne
- Inflammatory Bowel Disease Clinic, Division of Gastroenterology and Hepatology of Gastroenterology, University of Calgary, Calgary, Alberta, Canada
| | - Levinus A Dieleman
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Mark S Silverberg
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, University of Toronto Temerty Faculty of Medicine, Toronto, Ontario, Canada
| | - A Hillary Steinhart
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, University of Toronto Temerty Faculty of Medicine, Toronto, Ontario, Canada
| | - Paul Moayyedi
- Department of Medicine, Farncombe Family Digestive Health Research Institute McMaster University, Hamilton, Ontario, Canada
| | - Dan Turner
- The Juliet Keidan Institute of Pediatric Gastroenterology and Nutrition, The Hebrew University of Jerusalem, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Anne M Griffiths
- Department of Gastroenterology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, University of Toronto Temerty Faculty of Medicine, Toronto, Ontario, Canada
| | - Sun-Ho Lee
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, University of Toronto Temerty Faculty of Medicine, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, University of Toronto Temerty Faculty of Medicine, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Lalli MK, Salo TE, Hakola L, Knip M, Virtanen SM, Vatanen T. Associations between dietary fibers and gut microbiome composition in the EDIA longitudinal infant cohort. Am J Clin Nutr 2025; 121:83-99. [PMID: 39551356 DOI: 10.1016/j.ajcnut.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND The infant gut microbiome undergoes rapid changes in the first year of life, supporting normal development and long-term health. Although diet shapes this process, the role of fibers in complementary foods on gut microbiome maturation is poorly understood. OBJECTIVES We explored how the transition from human milk to fibers in complementary foods shapes the taxonomic and functional maturation of the gut microbiome within the first year of life. METHODS We assessed the longitudinal and cross-sectional development of infant gut microbiomes (N = 68 infants) and metabolomes (N = 33 infants) using linear mixed models to uncover their associations to dietary fibers and their food sources. Fiber intakes were assessed with 3-d food records (months 3, 6, 9, and 12) relying on CODEX-compliant fiber fraction values, and questionnaires tracked the overall complementary food introduction. Bacterial species were identified and quantified via MetaPhlAn2 from metagenomic data, and metabolomic profiles were obtained using 4 LC-MS methods. RESULTS We identified 176 complementary food fiber-bacterial species associations. First plant-based fibers associated with microbiota compositions similar to breastfeeding, and further associated with aromatic amino acid-derived metabolites, including 5-hydroxyindoleacetic acid (total dietary fiber - complementary foods (g) - β = 3.50, CI: 2.48, 4.52, P = 6.53 × 10-5). Distinct fibers from different food categories showed unique associations with specific bacterial taxa. Key species, such as Faecalibacterium prausnitznii, associated with oat fibers (g/MJ, β = 2.18, confidence interval: 1.36, 2.84, P = 6.12 × 10-6), reflective of maturing microbial communities. Fiber intake during weaning associated with shifts in metabolite profiles, including immunomodulatory metabolites, with fiber effects observed in a source- and timing-dependent manner, implicated in gradual microbiome diversification. CONCLUSIONS Introducing complementary dietary fibers during the weaning period supports gut microbiome diversification and stabilization. Even minor dietary variations shows significant associations with microbial taxa and functions from the onset of weaning, highlighting the importance of infant dietary recommendations that support the gut microbiome maturation during early life. This trial was registered at clinicaltrials.gov as registration number NCT01735123.
Collapse
Affiliation(s)
- Marianne K Lalli
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tuuli Ei Salo
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Leena Hakola
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Helsinki University Hospital, Helsinki, Finland; Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Suvi M Virtanen
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland; Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Tommi Vatanen
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, Cambridge, MA, United States; Liggins Institute, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
8
|
Ezzat L, Peter H, Bourquin M, Busi SB, Michoud G, Fodelianakis S, Kohler TJ, Lamy T, Geers A, Pramateftaki P, Baier F, Marasco R, Daffonchio D, Deluigi N, Wilmes P, Styllas M, Schön M, Tolosano M, De Staercke V, Battin TJ. Diversity and biogeography of the bacterial microbiome in glacier-fed streams. Nature 2025:10.1038/s41586-024-08313-z. [PMID: 39743584 DOI: 10.1038/s41586-024-08313-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 10/30/2024] [Indexed: 01/04/2025]
Abstract
The rapid melting of mountain glaciers and the vanishing of their streams is emblematic of climate change1,2. Glacier-fed streams (GFSs) are cold, oligotrophic and unstable ecosystems in which life is dominated by microbial biofilms2,3. However, current knowledge on the GFS microbiome is scarce4,5, precluding an understanding of its response to glacier shrinkage. Here, by leveraging metabarcoding and metagenomics, we provide a comprehensive survey of bacteria in the benthic microbiome across 152 GFSs draining the Earth's major mountain ranges. We find that the GFS bacterial microbiome is taxonomically and functionally distinct from other cryospheric microbiomes. GFS bacteria are diverse, with more than half being specific to a given mountain range, some unique to single GFSs and a few cosmopolitan and abundant. We show how geographic isolation and environmental selection shape their biogeography, which is characterized by distinct compositional patterns between mountain ranges and hemispheres. Phylogenetic analyses furthermore uncovered microdiverse clades resulting from environmental selection, probably promoting functional resilience and contributing to GFS bacterial biodiversity and biogeography. Climate-induced glacier shrinkage puts this unique microbiome at risk. Our study provides a global reference for future climate-change microbiology studies on the vanishing GFS ecosystem.
Collapse
Affiliation(s)
- Leïla Ezzat
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland.
- MARBEC, University of Montpellier, CNRS, IFREMER, IRD, Montpellier, France.
| | - Hannes Peter
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Massimo Bourquin
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Susheel Bhanu Busi
- UK Centre for Ecology and Hydrology, Wallingford, UK
- Systems Ecology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Grégoire Michoud
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Stilianos Fodelianakis
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Tyler J Kohler
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
- Department of Ecology, Faculty of Science, Charles University, Prague, Czechia
| | - Thomas Lamy
- MARBEC, University of Montpellier, CNRS, IFREMER, IRD, Montpellier, France
| | - Aileen Geers
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Paraskevi Pramateftaki
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Florian Baier
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Ramona Marasco
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Daniele Daffonchio
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Nicola Deluigi
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Paul Wilmes
- Systems Ecology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michail Styllas
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
- Institut de Physique du Globe de Paris, Paris, France
| | - Martina Schön
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Matteo Tolosano
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Vincent De Staercke
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland
| | - Tom J Battin
- River Ecosystems Laboratory, Alpine and Polar Environmental Research Center, Ecole Polytechnique Fédérale de Lausanne, Sion, Switzerland.
| |
Collapse
|
9
|
Wyatt NJ, Watson H, Young GR, Doona M, Tilling N, Allerton D, Masi AC, Ahmad T, Doyle JA, Frith K, Hart A, Hildreth V, Irving PM, Jones C, Kennedy NA, Lawrence S, Lees CW, Lees R, Liddle T, Lindsay JO, Marchesi JR, Parkes M, Powell N, Prescott NJ, Raine T, Satsangi J, Whelan K, Wood R, King A, Jostins-Dean L, Speight RA, McGregor N, Stewart CJ, Lamb CA. Evaluation of intestinal biopsy tissue preservation methods to facilitate large-scale mucosal microbiota research. EBioMedicine 2024; 112:105550. [PMID: 39742562 DOI: 10.1016/j.ebiom.2024.105550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND Large-scale multicentre studies are needed to understand complex relationships between the gut microbiota, health and disease. Interrogating the mucosal microbiota may identify important biology not captured by stool analysis. Gold standard tissue cryopreservation ('flash freezing') limits large-scale study feasibility. We aimed to compare gut microbiota in gold standard and pragmatic mucosal biopsy storage conditions. METHODS We collected endoscopic recto-sigmoid biopsies from 20 adults with inflammatory bowel disease. Biopsies were preserved using three methods: (i) flash freezing (most proximal and distal biopsy sites); (ii) nucleic acid preservative reagents (QIAGEN Allprotect®, Invitrogen RNAlater™, and Zymo DNA/RNA Shield™); and (iii) formalin fixation with paraffin embedding (FFPE), which is used to preserve tissue for clinical histopathology within healthcare settings. Microbiota were sequenced on the MiSeq platform (V4 region, 16S rRNA gene). FINDINGS Tissue microbiota were consistent between most proximal and distal tissue suggesting any within-patient variation observed reflected storage condition, not biopsy location. There was no significant difference in alpha-diversity or microbial community profiles of reagent-preserved versus gold standard tissue. FFPE community structure was significantly dissimilar to other tissue samples, driven by differential relative abundance of obligate gut anaerobes; Faecalibacterium, Anaerostipes and Lachnospiraceae. Despite these differences, tissue microbiota grouped by participant regardless of preservation and storage conditions. INTERPRETATION Preservative reagents offer a convenient alternative to flash freezing tissue in prospective large-scale mucosal microbiota studies. Whilst less comparable, FFPE provides potential for retrospective microbiota studies using historical samples. FUNDING Medical Research Council (MR/T032162/1) and The Leona M. and Harry B. Helmsley Charitable Trust (G-2002-04255).
Collapse
Affiliation(s)
- Nicola J Wyatt
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Hannah Watson
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Gregory R Young
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Mary Doona
- Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Ned Tilling
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dean Allerton
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrea C Masi
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Tariq Ahmad
- Department of Gastroenterology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom; Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, United Kingdom
| | - Jennifer A Doyle
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Katherine Frith
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ailsa Hart
- Department of Gastroenterology, St Marks Hospital and Academic Institute, Gastroenterology, London, United Kingdom; Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Victoria Hildreth
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Peter M Irving
- Department of Gastroenterology, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom; School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Claire Jones
- Department of Histopathology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Nicholas A Kennedy
- Department of Gastroenterology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom; Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, United Kingdom
| | - Sarah Lawrence
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Charlie W Lees
- Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom; Edinburgh IBD Unit, Western General Hospital, NHS Lothian, Edinburgh, United Kingdom
| | - Robert Lees
- Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Trevor Liddle
- Research Informatics Team, Clinical Research, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - James O Lindsay
- Department of Gastroenterology, Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom; Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Julian R Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, St Mary's Hospital, Imperial College London, London, United Kingdom
| | - Miles Parkes
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Nick Powell
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, St Mary's Hospital, Imperial College London, London, United Kingdom; Department of Gastroenterology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Natalie J Prescott
- Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, London, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Jack Satsangi
- Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, United Kingdom
| | - Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, United Kingdom
| | - Ruth Wood
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew King
- Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Luke Jostins-Dean
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - R Alexander Speight
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Naomi McGregor
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher J Stewart
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher A Lamb
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
10
|
Nishio J, Sato H, Watanabe E, Masuoka H, Aoki K, Kawazoe M, Wakiya R, Yamada S, Muraoka S, Masuoka S, Hayashi T, Mizutani S, Yamada Z, Koshiba K, Irita I, Kanaji M, Furukawa K, Yajima N, Dobashi H, Hirose W, Ishii Y, Suda W, Nanki T. Associations of gut microbiota with disease development, disease activity, and therapeutic effects in patients with systemic lupus erythematosus. Sci Rep 2024; 14:32076. [PMID: 39738678 PMCID: PMC11685445 DOI: 10.1038/s41598-024-83835-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
Altered gut microbiota is linked to systemic lupus erythematosus (SLE), but its association with disease development, disease activity, and post-intervention changes remains unclear. We compared new-onset SLE (NOSLE, n = 25), SLE in remission (RemSLE, n = 30), and healthy controls (HC, n = 30) cross-sectionally and conducted the first longitudinal analysis of NOSLE patients (n = 22) from pre-intervention to remission over 12 months. Significant β-diversity differences were observed in both NOSLE and RemSLE compared to HC, but not between NOSLE and RemSLE. Only four operational taxonomic units (OTUs) were enriched in NOSLE versus HC. However, 26 OTUs, including butyrate-producing bacteria (BPB), were depleted, and seven (including five BPBs) remained depleted in RemSLE compared to HC. OTUs positively and negatively correlated with disease activity were also identified. Longitudinal analysis revealed a reversal of several OTUs altered at onset and an increase in Streptococci, unrelated to the disease. Significant β-diversity differences were observed in patients with anti-SSA or anti-RNP antibodies and those with complement reduction compared to their counterparts. Our study identified gut microbiota alterations, including BPB depletion, in SLE regardless of onset or remission status, bacteria linked to disease activity, and a reversal of disease-associated bacteria along with the enrichment of Streptococci through intervention.
Collapse
Affiliation(s)
- Junko Nishio
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
- Department of Immunopathology and Immunoregulation, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Hiroshi Sato
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Eri Watanabe
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Hiroaki Masuoka
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Kotaro Aoki
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Mai Kawazoe
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Risa Wakiya
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa Prefecture, 761-0793, Japan
| | - Soichi Yamada
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Sei Muraoka
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Shotaro Masuoka
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Tomoki Hayashi
- Division of Rheumatology, Department of Medicine, Showa University, 2-14-19 Nishinakanobu, Shinagawa-ku, Tokyo, 142-0054, Japan
| | - Satoshi Mizutani
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Zento Yamada
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Keiko Koshiba
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Izumi Irita
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Miwa Kanaji
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Karin Furukawa
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Nobuyuki Yajima
- Division of Rheumatology, Department of Medicine, Showa University, 2-14-19 Nishinakanobu, Shinagawa-ku, Tokyo, 142-0054, Japan
| | - Hiroaki Dobashi
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa Prefecture, 761-0793, Japan
| | - Wataru Hirose
- Hirose Clinic of Rheumatology, 2-14-7 Midoricho, Tokorozawa City, Saitama, 359-1111, Japan
| | - Yoshikazu Ishii
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Wataru Suda
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Toshihiro Nanki
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan.
| |
Collapse
|
11
|
Reigel AM, Easson CG, Fiore CL, Apprill A. Sponge exhalent metabolites influence coral reef picoplankton dynamics. Sci Rep 2024; 14:31394. [PMID: 39733106 DOI: 10.1038/s41598-024-82995-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
Coral reef sponges efficiently take up particulate and dissolved organic matter (DOM) from the water column and release compounds such as nucleosides, amino acids, and other dissolved metabolites to the surrounding reef via their exhalent seawater, but the influence of this process on reef picoplankton and nutrient processing is relatively unexplored. Here we examined the impact of sponge exhalent on the reef picoplankon community and subsequent alterations to the reef dissolved metabolite pool. We exposed reef picoplankton communities to a sponge exhalent water mixture (Niphates digitalis and Xestospongia muta) or filtered reef seawater (control) in closed, container-based dark incubations. We used 16S rRNA gene sequencing and flow cytometry-based cell counts to examine the picoplankton community and metabolomics and other analyses to examine the dissolved metabolite pool. The initial sponge exhalent was enriched in adenosine, inosine, chorismate, humic-like and amino acid-like components, and ammonium. Following 48 h of exposure to sponge exhalent, the picoplankton differed in composition, were reduced in diversity, showed doubled (or higher) growth efficiencies, and harbored increased copiotrophic and denitrifying taxa (Marinomonas, Pontibacterium, Aliiroseovarius) compared to control, reef-water based incubations. Alongside these picoplankton alterations, the sponge treatments, relative to seawater controls, had decreased adenosine, inosine, tryptophan, and ammonium, metabolites that may support the observed higher picoplankton growth efficiencies. Sponge treatments also had a net increase in several monosaccharides and other metabolites including anthranilate, riboflavin, nitrite, and nitrate. Our work demonstrates a link between sponge exhalent-associated metabolites and the picoplankton community, with exhalent water supporting an increased abundance of efficient, copiotrophic taxa that catabolize complex nutrients. The copiotrophic taxa were often different from those observed in previous algae and coral studies. These results have implications for better understanding the multifaceted role of sponges on picoplankton biomass with subsequent potential impacts to coral and other planktonic feeders in oligotrophic reef environments.
Collapse
Affiliation(s)
- Alicia M Reigel
- Department of Biology, Appalachian State University, Boone, USA.
- Biology Department, Washington and Lee University, Lexington, VA, 24450, USA.
| | - Cole G Easson
- Department of Biology, Middle Tennessee State University, Murfreesboro, USA
| | - Cara L Fiore
- Department of Biology, Appalachian State University, Boone, USA
| | - Amy Apprill
- Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Falmouth, USA
| |
Collapse
|
12
|
Bao Z, Yang Z, Sun R, Chen G, Meng R, Wu W, Li MD. Predicting host health status through an integrated machine learning framework: insights from healthy gut microbiome aging trajectory. Sci Rep 2024; 14:31143. [PMID: 39732755 DOI: 10.1038/s41598-024-82418-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
The gut microbiome, recognized as a critical component in the development of chronic diseases and aging processes, constitutes a promising approach for predicting host health status. Previous research has underscored the potential of microbiome-based predictions, and the rapid advancements of machine learning techniques have introduced new opportunities for exploiting microbiome data. To predict various host nonhealthy conditions, this study proposed an integrated machine learning-based estimation pipeline of Gut Age Index (GAI) by establishing a health aging baseline with the gut microbiome data from healthy individuals. We assessed the performance of GAI pipeline on two extensive cohorts - the Guangdong Gut Microbiome Project (GGMP) and the American Gut Project (AGP). In the GGMP cohort, for 20 common chronic diseases such as metabolic syndrome, obesity, and cardiovascular diseases, the proposed GAI achieved a balanced accuracy, ranging from 66 to 75%, with the prediction performance for atherosclerosis being the highest. In the AGP cohort, the balanced accuracy of GAI ranged from 58 to 72% for 10 diseases. Based on the results from these two datasets, we conclude that our proposed approach in this study can be used to predict individual health status, which offers the potential for scalable, cost-effective, and personalized health insights.
Collapse
Affiliation(s)
- Zhiwei Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruixiang Sun
- The Maiyata Research Institute For Beneficial Bacteria, Shaoxing, Zhejiang, China
| | - Guoliang Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiling Meng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Wei Wu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China.
- Guangdong Provincial Institute of Public Health, Guangzhou, China.
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, China.
| |
Collapse
|
13
|
Bisker C, Taylor G, Carney H, Orr CH, Javan GT, Ralebitso-Senior TK. Comparative soil bacterial metabarcoding after aboveground vs. subsurface decomposition of Mus musculus. Sci Rep 2024; 14:31179. [PMID: 39732855 DOI: 10.1038/s41598-024-82437-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
Outdoor microcosms, metabarcoding with next-generation sequencing of the 16S rRNA bacterial gene, total body score (TBS) and physicochemical analyses were used to monitor Mus musculus decomposition aboveground (A) and in the subsurface (S), and compared to soil-only controls (C). As determined by MaAsLin2 analysis, significant shifts in bacterial communities at 30 cm depths within the A, S and C treatments distinguished control from experimental soils, and between aboveground and subsurface deposition, demonstrating the potential for gravesoil discrimination during the first 90 days. For example, Dokdonella (p = 0.0002), Edaphobaculum (p = 0.0004) and Lacibacter (p = 0.0034) recorded significant shifts relative to sampling time. Furthermore, Massilia (p = 0.0005), Mycobacterium (p = 0.0006) and Sandaracinus (p = 0.0007) increased in abundance for the aboveground mice treatments. This was confirmed with ANOSIM where p = 0.0082 showed statistically significant difference between the aboveground and subsurface deposition. TBS and physicochemical analyses suggested that nutrient release into the soils occurred during active decay and skin rapture on days 7-13 in the subsurface and days 13-20 aboveground, with a particular increase in soil potassium concentration on day 15. Significant differences in soil temperatures resulted between A and S vs. C microcosms, aligning with atmospheric temperature changes. In summary, complementary application of metabarcoding, total body score, exogenous and physicochemical methods for postmortem interval estimation and clandestine grave location highlighted the feasibility of using temperature records downloaded from meteorological stations and portable X-ray fluorescence as indicators for various phases of decomposition.
Collapse
Affiliation(s)
- Chawki Bisker
- National Institute of Criminalistics and Criminology, Bouchaoui, Algiers, Algeria
- School of Health and Life Sciences, Teesside University, Middlesbrough, UK
| | - Gillian Taylor
- School of Health and Life Sciences, Teesside University, Middlesbrough, UK
- National Horizons Centre, Darlington, UK
| | - Helen Carney
- School of Health and Life Sciences, Teesside University, Middlesbrough, UK
- National Horizons Centre, Darlington, UK
| | - Caroline H Orr
- School of Health and Life Sciences, Teesside University, Middlesbrough, UK
- National Horizons Centre, Darlington, UK
| | - Gulnaz T Javan
- Department of Physical Sciences and Forensic Science Programs, Alabama State University, Montgomery, AL, USA
| | - Theresia Komang Ralebitso-Senior
- School of Health and Life Sciences, Teesside University, Middlesbrough, UK.
- School of Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
14
|
Kim CY, Park DJ, Ahn BC, Baek S, Hong MH, Nguyen LT, Hwang SH, Kim N, Podlesny D, Orakov A, Schudoma C, Robbani SM, Shim HS, Yoon HI, Lee CY, Park SY, Yong D, Han M, Bork P, Kim BC, Ha SJ, Kim HR, Lee I. A conserved pilin from uncultured gut bacterial clade TANB77 enhances cancer immunotherapy. Nat Commun 2024; 15:10726. [PMID: 39730328 DOI: 10.1038/s41467-024-55388-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/10/2024] [Indexed: 12/29/2024] Open
Abstract
Immune checkpoint blockade (ICB) has become a standard anti-cancer treatment, offering durable clinical benefits. However, the limited response rate of ICB necessitates biomarkers to predict and modulate the efficacy of the therapy. The gut microbiome's influence on ICB efficacy is of particular interest due to its modifiability through various interventions. However, gut microbiome biomarkers for ICB response have been inconsistent across different studies. Here, we identify TANB77, an uncultured and distinct bacterial clade, as the most consistent responder-enriched taxon through meta-analysis of ten independent ICB recipient cohorts. Traditional taxonomy fails to distinguish TANB77 from unrelated taxa, leading to its oversight. Mice with higher gut TANB77 abundance, either naturally or through transplantation, show improved response to anti-PD-1 therapy. Additionally, mice injected with TANB77-derived pilin-like protein exhibit improved anti-PD-1 therapy response, providing in vivo evidence for the beneficial role of the pilin-like protein. These findings suggest that pilins from the TANB77 order may enhance responses to ICB therapy across diverse cohorts of cancer patients.
Collapse
Affiliation(s)
- Chan Yeong Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, 69117, Heidelberg, Germany
| | - Dong Jin Park
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Beung Chul Ahn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Center for Lung Cancer, Division of Hematology and Oncology, Department of Internal Medicine, Research Institute and Hospital, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Seungbyn Baek
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea
| | - Min Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Linh Thanh Nguyen
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Sun Ha Hwang
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Nayeon Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea
| | - Daniel Podlesny
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, 69117, Heidelberg, Germany
| | - Askarbek Orakov
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, 69117, Heidelberg, Germany
| | - Christian Schudoma
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, 69117, Heidelberg, Germany
| | - Shahriyar Mahdi Robbani
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, 69117, Heidelberg, Germany
| | - Hyo Sup Shim
- Department of Pathology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hong In Yoon
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Chang Young Lee
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seong Yong Park
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dongeun Yong
- Department of Laboratory Medicine, Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Mina Han
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Peer Bork
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, 69117, Heidelberg, Germany
| | - Byoung Choul Kim
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea.
| | - Sang-Jun Ha
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea.
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea.
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea.
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea.
| |
Collapse
|
15
|
Alhasani AT, Modasia AA, Anodiyil M, Corsetti M, Aliyu AI, Crooks C, Marciani L, Reid J, Yakubov GE, Avery A, Harris H, Warren FJ, Spiller RC. Mode of Action of Psyllium in Reducing Gas Production from Inulin and its Interaction with Colonic Microbiota: A 24 hours, Randomised, Placebo-Controlled Trial in Healthy Human Volunteers. J Nutr 2024:S0022-3166(24)01244-6. [PMID: 39732438 DOI: 10.1016/j.tjnut.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Recent studies show that the increase in breath hydrogen (BH2) and symptoms after ingestion of inulin is reduced by co-administering psyllium. OBJECTIVES To determine if slowing delivery of inulin to the colon by administering it in divided doses would mimic the effect of psyllium. Primary endpoint was the BH2 area under the curve AUC0-24hours. Secondary endpoints included BH2 AUC0-6h, 6-12h, and 12-24h. Exploratory endpoints included the correlation of BH2 AUC0-24h with dietary FODMAPs intake and in vitro fermentation results. METHODS 17 Healthy adults were randomised to a single-blind, 3-arm, crossover trial. All consumed 20g inulin (I) powder dissolved in 500mL water and mixed with either 20g maltodextrin (control) or 20g psyllium (PI) consumed as a single dose or 20g inulin given in divided doses (DDI), 62.5mL every 45 minutes over 6h. 24-h BH2, dietary FODMAP intake, stool microbiota, and gas production in vitro were measured. Responders were defined as those whose AUC0-24h BH2 was reduced by psyllium, while non-responders showed no reduction. RESULTS Compared to control, PI did not reduce average BH2 AUC0-24h while DDI increased it, p<0.0002. DDI and PI both significantly reduced BH2 AUC0-6h compared to the control, p<0.0001. However, subsequently, DDI significantly increased BH2 from 6-12h (p<0.0001) and overnight (12-24h) (p<0.0001), while PI did so only overnight (p=0.0002). Non-responders showed greater release of arabinose during in-vitro fermentation and higher abundance of two species, Clostridium spp. AM22_11AC and Phocaeicola dorei, which also correlated with BH2 production on PI. Dietary FODMAP intake tended to correlate inversely with BH2 AUC0-24h (r=-0.42, p=0.09) and correlated with microbiome community composition. CONCLUSIONS DDI, like psyllium, reduces early BH2 production. Psyllium acts by delaying transit to the colon but not reducing colonic fermentation over 24h. Dietary FODMAP intake correlates with BH2 response to inulin and the microbiome. CLINICAL TRIAL REGISTRY NUMBER www. CLINICALTRIALS govID: NCT05619341Ethical approval for this study was obtained from the Research Ethics Committee at the Faculty of Medicine and Health Sciences, University of Nottingham (FMHS 17-622). STATEMENT OF SIGNIFICANCE This mechanistic study adds to recent evidence that psyllium reduces colonic gas and symptoms in the 6 hours after ingestion. The underlying mechanism includes slowing of delivery to the colon, but total fermentation over 24 hours is not altered. While some subjects do show a reduction, not all do, possibly due to the degradation of psyllium by the colonic microbiota. Habitual FODMAP intake correlates negatively with gas production, raising the possibility that dietary manipulation could alter colonic fermentation pathways to produce less gas.
Collapse
Affiliation(s)
- Alaa T Alhasani
- Nottingham NIHR Biomedical Research Centre and Nottingham Digestive Disease Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom; Faculty of Health and Rehabilitation Sciences, Princess Nourah Bint Abdul Rahman University, Riyadh, Saudi Arabia
| | - Amisha A Modasia
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Mohamed Anodiyil
- Nottingham NIHR Biomedical Research Centre and Nottingham Digestive Disease Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Maura Corsetti
- Nottingham NIHR Biomedical Research Centre and Nottingham Digestive Disease Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Abdulsalam I Aliyu
- Nottingham NIHR Biomedical Research Centre and Nottingham Digestive Disease Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Colin Crooks
- Nottingham NIHR Biomedical Research Centre and Nottingham Digestive Disease Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Luca Marciani
- Nottingham NIHR Biomedical Research Centre and Nottingham Digestive Disease Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Joshua Reid
- Food and Biomaterials Laboratory, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Gleb E Yakubov
- Food and Biomaterials Laboratory, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Amanda Avery
- Food and Biomaterials Laboratory, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Hannah Harris
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Frederick J Warren
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Robin C Spiller
- Nottingham NIHR Biomedical Research Centre and Nottingham Digestive Disease Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
16
|
Huang KD, Müller M, Sivapornnukul P, Bielecka AA, Amend L, Tawk C, Lesker TR, Hahn A, Strowig T. Dietary selective effects manifest in the human gut microbiota from species composition to strain genetic makeup. Cell Rep 2024; 43:115067. [PMID: 39673707 DOI: 10.1016/j.celrep.2024.115067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 12/16/2024] Open
Abstract
Diet significantly influences the human gut microbiota, a key player in health. We analyzed shotgun metagenomic sequencing data from healthy individuals with long-term dietary patterns-vegan, flexitarian, or omnivore-and included detailed dietary surveys and blood biomarkers. Dietary patterns notably affected the bacterial community composition by altering the relative abundances of certain species but had a minimal impact on microbial functional repertoires. However, diet influenced microbial functionality at the strain level, with diet type linked to strain genetic variations. We also found molecular signatures of selective pressure in species enriched by specific diets. Notably, species enriched in omnivores exhibited stronger positive selection, such as multiple iron-regulating genes in the meat-favoring bacterium Odoribacter splanchnicus, an effect that was also validated in independent cohorts. Our findings offer insights into how diet shapes species and genetic diversity in the human gut microbiota.
Collapse
Affiliation(s)
- Kun D Huang
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Mattea Müller
- Institute of Food Science and Nutrition, Leibniz University of Hannover, Hannover, Germany
| | - Pavaret Sivapornnukul
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany; Center of Excellence in Systems Microbiology (CESM), Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Agata Anna Bielecka
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lena Amend
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Caroline Tawk
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Till-Robin Lesker
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Hahn
- Institute of Food Science and Nutrition, Leibniz University of Hannover, Hannover, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany; Hannover Medical School (MHH), Hannover, Germany; Centre for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|
17
|
Bhosle A, Jackson MI, Walsh AM, Franzosa EA, Badri DV, Huttenhower C. Response of the gut microbiome and metabolome to dietary fiber in healthy dogs. mSystems 2024:e0045224. [PMID: 39714168 DOI: 10.1128/msystems.00452-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/21/2024] [Indexed: 12/24/2024] Open
Abstract
Dietary fiber confers multiple health benefits originating from the expansion of beneficial gut microbial activity. However, very few studies have established the metabolic consequences of interactions among specific fibers, microbiome composition, and function in either human or representative animal models. In a study design reflective of realistic population dietary variation, fecal metagenomic and metabolomic profiles were analyzed from healthy dogs fed 12 test foods containing different fiber sources and quantities (5-13% as-fed basis). Taxa and functions were identified whose abundances were associated either with overall fiber intake or with specific fiber compositions. Fourteen microbial species were significantly enriched in response to ≥1 specific fiber source; enrichment of fiber-derived metabolites was more pronounced in response to these fiber sources. Positively associated fecal metabolites, including short-chain fatty acids, acylglycerols, fiber bound sugars, and polyphenols, co-occurred with microbes enriched in specific food groups. Critically, the specific metabolite pools responsive to differential fiber intake were dependent on differences both in individual microbial community membership and in overall ecological configuration. This helps to explain, for the first time, differences in microbiome-diet associations observed in companion animal epidemiology. Thus, our study corroborates findings in human cohorts and reinforces the role of personalized microbiomes even in seemingly phenotypically homogeneous subjects. IMPORTANCE Consumption of dietary fiber changes the composition of the gut microbiome and, to a larger extent, the associated metabolites. Production of health-relevant metabolites such as short-chain fatty acids from fiber depends both on the consumption of a specific fiber and on the enrichment of beneficial metabolite-producing species in response to it. Even in a seemingly homogeneous population, the benefit received from fiber consumption is personalized and emphasizes specific fiber-microbe-host interactions. These observations are relevant for both population-wide and personalized nutrition applications.
Collapse
Affiliation(s)
- Amrisha Bhosle
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Aaron M Walsh
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Eric A Franzosa
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Curtis Huttenhower
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Ruff SE, de Angelis IH, Mullis M, Payet JP, Magnabosco C, Lloyd KG, Sheik CS, Steen AD, Shipunova A, Morozov A, Reese BK, Bradley JA, Lemonnier C, Schrenk MO, Joye SB, Huber JA, Probst AJ, Morrison HG, Sogin ML, Ladau J, Colwell F. A global comparison of surface and subsurface microbiomes reveals large-scale biodiversity gradients, and a marine-terrestrial divide. SCIENCE ADVANCES 2024; 10:eadq0645. [PMID: 39693444 DOI: 10.1126/sciadv.adq0645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024]
Abstract
Subsurface environments are among Earth's largest habitats for microbial life. Yet, until recently, we lacked adequate data to accurately differentiate between globally distributed marine and terrestrial surface and subsurface microbiomes. Here, we analyzed 478 archaeal and 964 bacterial metabarcoding datasets and 147 metagenomes from diverse and widely distributed environments. Microbial diversity is similar in marine and terrestrial microbiomes at local to global scales. However, community composition greatly differs between sea and land, corroborating a phylogenetic divide that mirrors patterns in plant and animal diversity. In contrast, community composition overlaps between surface to subsurface environments supporting a diversity continuum rather than a discrete subsurface biosphere. Differences in microbial life thus seem greater between land and sea than between surface and subsurface. Diversity of terrestrial microbiomes decreases with depth, while marine subsurface diversity and phylogenetic distance to cultured isolates rivals or exceeds that of surface environments. We identify distinct microbial community compositions but similar microbial diversity for Earth's subsurface and surface environments.
Collapse
Affiliation(s)
- S Emil Ruff
- Marine Biological Laboratory, Woods Hole, MA, USA
| | | | | | - Jérôme P Payet
- College of Earth, Ocean, and Atmospheric Sciences, Oregon State University, Corvallis, OR, USA
| | | | | | - Cody S Sheik
- Large Lakes Observatory and Department of Biology, University of Minnesota Duluth, Duluth, MN, USA
| | | | | | | | - Brandi Kiel Reese
- University of South Alabama, Mobile, AL, USA
- Dauphin Island Sea Laboratory, Dauphin Island, AL, USA
| | - James A Bradley
- Aix Marseille University, University of Toulon, CNRS, IRD, MIO, Marseille, France
- Queen Mary University of London, London, UK
| | - Clarisse Lemonnier
- UMR CARRTEL, INRAE, Université Savoie Mont-Blanc, Thonon-les-Bains, France
| | - Matthew O Schrenk
- Department of Earth and Environmental Sciences, Michigan State University, East Lansing, MI. USA
| | - Samantha B Joye
- Department of Marine Sciences, University of Georgia, Athens, GA, USA
| | - Julie A Huber
- Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - Alexander J Probst
- Environmental Metagenomics, Research Center One Health Ruhr of the University Alliance Ruhr, Faculty of Chemistry and Centre of Water and Environmental Research (ZWU), University of Duisburg-Essen, Essen, Germany
| | | | | | - Joshua Ladau
- Department of Computational Precision Health, University of California, San Francisco, CA, USA
- Arva Intelligence, Houston, TX, USA
| | - Frederick Colwell
- College of Earth, Ocean, and Atmospheric Sciences, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
19
|
He B, Xu S, Schooling CM, Leung GM, Ho JWK, Au Yeung SL. Gut microbiome and obesity in late adolescence: A case-control study in "Children of 1997" birth cohort. Ann Epidemiol 2024; 101:58-66. [PMID: 39710013 DOI: 10.1016/j.annepidem.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
PURPOSE Although the gut microbiome is important in human health, its relation to adolescent obesity remains unclear. Here we assessed the associations of the gut microbiome with adolescent obesity in a case-control study. METHODS In the "Children of 1997" birth cohort, participants with and without obesity at ∼17.4 years were 1:1 matched on sex, physical activity, parental education and occupation (n = 312). Fecal gut microbiome composition and pathways were assessed via shotgun metagenomic sequencing. The association of microbiota species with obesity was evaluated using conditional logistic regression. We explored the association of the obesity-relevant species with adolescent metabolomics using multivariable linear regression, and causal relationships with type 2 diabetes using Mendelian randomization analysis. RESULTS Gut microbiota in the adolescents with obesity exhibited lower richness (p = 0.031) and evenness (p = 0.014) compared to controls. Beta diversity revealed differences in the microbiome composition in two groups (p = 0.034). Lower relative abundance of Clostridium spiroforme, Clostridium phoceensis and Bacteroides uniformis were associated with higher obesity risk (q<0.15). Lower Bacteroides uniformis was associated with higher branched-chain amino acid, potentially contributing to higher type 2 diabetes risk. CONCLUSION Adolescents with obesity had a distinct gut microbiota profile compared to the controls, possibly linked to metabolic pertubation and related diseases.
Collapse
Affiliation(s)
- Baoting He
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| | - Sheng Xu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Laboratory of Data Discovery for Health Limited (D(2)4H), Hong Kong Science Park, Hong Kong.
| | - C Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Public Health and Health Policy, City University of New York, New York, USA.
| | - Gabriel M Leung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Laboratory of Data Discovery for Health Limited (D(2)4H), Hong Kong Science Park, Hong Kong.
| | - Joshua W K Ho
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Laboratory of Data Discovery for Health Limited (D(2)4H), Hong Kong Science Park, Hong Kong.
| | - Shiu Lun Au Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| |
Collapse
|
20
|
Jang J, Park J, Hwang CY, Gim Y, Park KT, Yoon YJ, Seo M, Lee BY. Selective transmission of airborne bacterial communities from the ocean to the atmosphere over the Northern Pacific Ocean. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177462. [PMID: 39528211 DOI: 10.1016/j.scitotenv.2024.177462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/24/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
This study simultaneously measured the taxonomic diversity of bacterial communities in both seawater and PM2.5 aerosol samples collected from the Northern Pacific Ocean during a cruise covering 7724 km between 37°N 126°E and 58°N 179°E. The relative abundance of Proteobacteria, Cyanobacteria, and Firmicutes were found to be more prevalent in aerosol samples (39 ± 16 %, 5.1 ± 1.9 %, and 3.2 ± 1.7 %, respectively) than in seawater samples (26 ± 9 %, 3.8 ± 1.7 %, and 0.02 ± 0.09 %, respectively). The preferential aerosolization of bacterial communities such as Proteobacteria and Firmicutes was likely to be accompanied by a terrestrial origin and high hydrophobicity. Cyanobacteria could undergo increased aerosolization, possibly because of their smaller size in the significantly higher salinity open ocean (32.8 ± 0.14 PSU) compared to those in lower salinity coastal areas (31.3 ± 1.4 PSU). The results of this study indicated that bacterial properties substantially affect their transfer from the ocean to the atmosphere, possibly influencing climate change and public health.
Collapse
Affiliation(s)
- Jiyi Jang
- Division of Ocean and Atmospheric Sciences, Korea Polar Research Institute (KOPRI), Incheon, 21990, South Korea
| | - Jiyeon Park
- Division of Ocean and Atmospheric Sciences, Korea Polar Research Institute (KOPRI), Incheon, 21990, South Korea.
| | - Chung Yeon Hwang
- School of Earth and Environmental Sciences and Research Institute of Oceanography, Seoul National University, Seoul 08826, South Korea
| | - Yeontae Gim
- Division of Ocean and Atmospheric Sciences, Korea Polar Research Institute (KOPRI), Incheon, 21990, South Korea
| | - Ki-Tae Park
- Department of Environmental Sciences and Biotechnology, Hallym University, Gangwon-do 24252, South Korea
| | - Young Jun Yoon
- Division of Ocean and Atmospheric Sciences, Korea Polar Research Institute (KOPRI), Incheon, 21990, South Korea
| | - Minju Seo
- Division of Ocean and Atmospheric Sciences, Korea Polar Research Institute (KOPRI), Incheon, 21990, South Korea; University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Bang Yong Lee
- Division of Ocean and Atmospheric Sciences, Korea Polar Research Institute (KOPRI), Incheon, 21990, South Korea
| |
Collapse
|
21
|
Jeong S, Tollison TS, Brochu H, Huntress I, Yount KS, Zheng X, Darville T, O'Connell CM, Peng X. Cervicovaginal microbial features predict Chlamydia trachomatis spread to the upper genital tract of infected women. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625070. [PMID: 39651251 PMCID: PMC11623589 DOI: 10.1101/2024.11.26.625070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
INTRODUCTION Chlamydia trachomatis (CT) infection can lead to pelvic inflammatory disease, infertility and other reproductive sequelae when it ascends to the upper genital tract. Factors including chlamydial burden, co-infection with other sexually-transmitted bacterial pathogens and oral contraceptive use influence risk for upper genital tract spread. Cervicovaginal microbiome composition influences CT susceptibility and we investigated if it contributes to spread by analyzing amplicon sequence variants (ASVs) derived from the V4 region of 16S rRNA genes in vaginal samples collected from women at high risk for CT infection and for whom endometrial infection had been determined. RESULTS Participants were classified as CT negative (CT-, n=77), CT positive at the cervix (Endo-, n=77), or CT positive at both cervix and endometrium (Endo+, n=66). Although we were unable to identify many significant differences between CT infected and uninfected women, differences in abundance of ASVs representing Lactobacillus iners and L. crispatus subspecies but not dominant lactobacilli were detected. Twelve informative ASVs predicted endometrial chlamydial infection (AUC=0.74), with CT ASV abundance emerging as a key predictor. We also observed a positive correlation between levels of cervically secreted cytokines previously associated with CT ascension and abundance of the informative ASVs. CONCLUSION Our findings suggest that vaginal microbial community members may influence chlamydial spread directly by nutrient limitation and/or disrupting endocervical epithelial integrity and indirectly by modulating pro-inflammatory signaling and/or homeostasis of adaptive immunity. Further investigation of these predictive microbial factors may lead to cervicovaginal microbiome biomarkers useful for identifying women at increased risk for disease.
Collapse
|
22
|
Byrne A, Diener C, Brown BP, Maust BS, Feng C, Alinde BL, Gibbons SM, Koch M, Gray CM, Jaspan HB, Nyangahu DD. Neonates exposed to HIV but uninfected exhibit an altered gut microbiota and inflammation associated with impaired breast milk antibody function. MICROBIOME 2024; 12:261. [PMID: 39707483 DOI: 10.1186/s40168-024-01973-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/11/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Infants exposed to HIV but uninfected have altered immune profiles which include heightened systemic inflammation. The mechanism(s) underlying this phenomenon is unknown. Here, we investigated differences in neonatal gut bacterial and viral microbiome and associations with inflammatory biomarkers in plasma. Further, we tested whether HIV exposure impacts antibody-microbiota binding in neonatal gut and whether antibodies in breast milk impact the growth of commensal bacteria. RESULTS Neonates exposed to HIV but uninfected (nHEU) exhibited altered gut bacteriome and virome compared to unexposed neonates (nHU). In addition, HIV exposure differentially impacted IgA-microbiota binding in neonates. The relative abundance of Blautia spp. in the whole stool or IgA-bound microbiota was positively associated with plasma concentrations of C-reactive protein. Finally, IgA from the breast milk of mothers living with HIV displayed a significantly lower ability to inhibit the growth of Blautia coccoides which was associated with inflammation in nHEU. CONCLUSION nHEU exhibits profound alterations in gut bacterial microbiota with a mild impact on the enteric DNA virome. Elevated inflammation in nHEU could be due to a lower capacity of breast milk IgA from mothers living with HIV to limit growth the of gut bacteria associated with inflammation. Video Abstract.
Collapse
Affiliation(s)
- Audrey Byrne
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Christian Diener
- Institute For Systems Biology, Seattle, WA, 98109, USA
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Bryan P Brown
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Colin Feng
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Berenice L Alinde
- Stellenbosch University, Cape Town, South Africa
- Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Sean M Gibbons
- Institute For Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
- eScience Institute, University of Washington, Seattle, WA, 98195, USA
| | - Meghan Koch
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98101, USA
- Department of Immunology, University of Washington, Seattle, WA, 98195, USA
| | - Clive M Gray
- Stellenbosch University, Cape Town, South Africa
- Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Heather B Jaspan
- Seattle Children's Research Institute, Seattle, WA, USA
- Division of Immunology, University of Cape Town, Cape Town, South Africa
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Donald D Nyangahu
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
- Department of Pharmacology, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA.
- Center for Advanced Biotechnology and Medicine, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA.
- Department of Human Pathology, Egerton University, Nakuru, Kenya.
| |
Collapse
|
23
|
Nowotny HF, Zheng T, Seiter TM, Ju J, Schneider H, Kroiss M, Sarkis AL, Sturm L, Britz V, Lechner A, Potzel AL, Kunz S, Bidlingmaier M, Neuhaus K, Gottschlich A, Kobold S, Reisch N, Schirmer M, Reincke M, Adolf C. Sex-dependent modulation of T and NK cells and gut microbiome by low sodium diet in patients with primary aldosteronism. Front Immunol 2024; 15:1428054. [PMID: 39749333 PMCID: PMC11693743 DOI: 10.3389/fimmu.2024.1428054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025] Open
Abstract
Background High dietary sodium intake is a major cardiovascular risk factor and adversely affects blood pressure control. Patients with primary aldosteronism (PA) are at increased cardiovascular risk, even after medical treatment, and high dietary sodium intake is common in these patients. Here, we analyze the impact of a moderate dietary sodium restriction on microbiome composition and immunophenotype in patients with PA. Methods Prospective two-stage clinical trial including two subgroups: 15 treatment-naive PA patients compared to matched normotensive controls; and 31 PA patients on mineralocorticoid receptor antagonist treatment before and three months after sodium restriction. Patients underwent blood pressure measurements, laboratory tests, analysis of peripheral blood mononuclear cells via flow cytometry and microbiome analysis. Results We observed a higher percentage of Tregs in treatment-naive PA patients (p = 0.0303), while the abundance of Bacteroides uniformis was higher in PA patients compared to normotensive controls (p = 0.00027) and the abundance of Lactobacillus species however was higher in the subgroup of normotensive controls (p = 0.0290). Sodium restriction was accompanied by a decrease in pro-inflammatory Tc17 cells in male patients (p = 0.0081, females p = 0.3274). Bacteroides uniformis abundance was higher in female patients (0.01230, p = 0.0016) and decreased upon sodium restriction (0.002309, p = 0.0068). Conclusion Dietary sodium restriction in patients with PA modulates the peripheral immune cell composition toward a less inflammatory phenotype. This suggests a potential mechanism by which sodium reduction modulates immune cell composition, leading to blood pressure reduction and positively impacting cardiovascular risk.
Collapse
Affiliation(s)
- Hanna F. Nowotny
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Tingting Zheng
- Chair of Translational Microbiome Data Integration, Technical University of Munich, Freising, Germany
| | | | - Jing Ju
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Holger Schneider
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Kroiss
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Anna-Lina Sarkis
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lisa Sturm
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Vera Britz
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Andreas Lechner
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Anne L. Potzel
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Physicians Association for Nutrition e.V, Munich, Germany
- CCG Type 2 Diabetes, Helmholtz Zentrum München, Munich, Germany
| | - Sonja Kunz
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Martin Bidlingmaier
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Adrian Gottschlich
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Hospital LMU, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Hospital LMU, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Munich, Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Nicole Reisch
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Melanie Schirmer
- Chair of Translational Microbiome Data Integration, Technical University of Munich, Freising, Germany
| | - Martin Reincke
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Christian Adolf
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
24
|
Fishbein SRS, DeVeaux AL, Khanna S, Ferreiro AL, Liao J, Agee W, Ning J, Mahmud B, Wallace MJ, Hink T, Reske KA, Cass C, Guruge J, Leekha S, Rengarajan S, Dubberke ER, Dantas G. Commensal-pathogen dynamics structure disease outcomes during Clostridioides difficile colonization. Cell Host Microbe 2024:S1931-3128(24)00447-5. [PMID: 39731916 DOI: 10.1016/j.chom.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/24/2024] [Accepted: 12/02/2024] [Indexed: 12/30/2024]
Abstract
Gastrointestinal colonization by Clostridioides difficile is common in healthcare settings and ranges in presentation from asymptomatic carriage to lethal C. difficile infection (CDI). We used a systems biology approach to investigate why patients colonized with C. difficile have a range of clinical outcomes. Microbiota humanization of germ-free mice with fecal samples from toxigenic C. difficile carriers revealed a spectrum of virulence among clinically prevalent clade 1 lineages and identified candidate taxa, including Blautia, as markers of stable colonization. Using gnotobiotic mice engrafted with defined human microbiota, we validated strain-specific CDI severity across clade 1 strains isolated from patients. Mice engrafted with a community broadly representative of colonized patients were protected from severe disease across all strains without suppression of C. difficile colonization. These results underline the capacity of gut community structure to attenuate a diversity of pathogenic strains without inhibiting colonization, providing insight into determinants of stable C. difficile carriage.
Collapse
Affiliation(s)
- Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Anna L DeVeaux
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sakshi Khanna
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Aura L Ferreiro
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - James Liao
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Wesley Agee
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bejan Mahmud
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Miranda J Wallace
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Tiffany Hink
- Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Kimberly A Reske
- Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Candice Cass
- Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Janaki Guruge
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Sidh Leekha
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sunaina Rengarajan
- Department of Medicine, Division of Dermatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Erik R Dubberke
- Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St Louis, St. Louis, MO, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
25
|
Galic I, Bez C, Bertani I, Venturi V, Stankovic N. Herbicide-treated soil as a reservoir of beneficial bacteria: microbiome analysis and PGP bioinoculants in maize. ENVIRONMENTAL MICROBIOME 2024; 19:107. [PMID: 39695885 DOI: 10.1186/s40793-024-00654-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Herbicides are integral to agricultural weed management but can adversely affect non-target organisms, soil health, and microbiome. We investigated the effects of herbicides on the total soil bacterial community composition using 16S rRNA gene amplicon community profiling. Further, we aimed to identify herbicide-tolerant bacteria with plant growth-promoting (PGP) capabilities as a mitigative strategy for these negative effects, thereby promoting sustainable agricultural practices. RESULTS A bacterial community analysis explored the effects of long-term S-metolachlor application on soil bacterial diversity, revealing that the herbicide's impact on microbial communities is less significant than the effects of temporal factors (summer vs. winter) or agricultural practices (continuous maize cultivation vs. maize-winter wheat rotation). Although S-metolachlor did not markedly alter the overall bacteriome structure in our environmental context, the application of enrichment techniques enabled the selection of genera such as Pseudomonas, Serratia, and Brucella, which were rare in metagenome analysis of soil samples. Strain isolation revealed a rich source of herbicide-tolerant PGP bacteria within the culturable microbiome fraction, termed the high herbicide concentration tolerant (HHCT) bacterial culture collection. Within the HHCT collection, we isolated 120 strains that demonstrated significant in vitro PGP and biocontrol potential, and soil quality improvement abilities. The most promising HHCT isolates were combined into three consortia, each exhibiting a comprehensive range of plant-beneficial traits. We evaluated the efficacy and persistence of these multi-strain consortia during 4-week in pot experiments on maize using both agronomic parameters and 16S rRNA gene community analysis assessing early-stage plant development, root colonization, and rhizosphere persistence. Notably, 7 out of 10 inoculated consortia partners successfully established themselves and persisted in the maize root microbiome without significantly altering host root biodiversity. Our results further evidenced that all three consortia positively impacted both seed germination and early-stage plant development, increasing shoot biomass by up to 47%. CONCLUSIONS Herbicide-treated soil bacterial community analysis revealed that integrative agricultural practices can suppress the effects of continuous S-metolachlor application on soil microbial diversity and stabilize microbiome fluctuations. The HHCT bacterial collection holds promise as a source of beneficial bacteria that promote plant fitness while maintaining herbicide tolerance.
Collapse
Affiliation(s)
- Ivana Galic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, Belgrade 152, 11042, Serbia
| | - Cristina Bez
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, 34149, Trieste, Italy
| | - Iris Bertani
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, 34149, Trieste, Italy
| | - Vittorio Venturi
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, 34149, Trieste, Italy
- African Genome Center, University Mohammed VI Polytechnic, Lot 660, Hay Moulay Rachid, 43150, Ben Guerir, Morocco
| | - Nada Stankovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, Belgrade 152, 11042, Serbia.
| |
Collapse
|
26
|
Gao F, Shen Y, Wu H, Laue HE, Lau FK, Gillet V, Lai Y, Shrubsole MJ, Prada D, Zhang W, Liu Z, Bellenger JP, Takser L, Baccarelli AA. Associations of Stool Metal Exposures with Childhood Gut Microbiome Multiomics Profiles in a Prospective Birth Cohort Study. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:22053-22063. [PMID: 39630952 DOI: 10.1021/acs.est.4c09642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Metal exposures are closely related to childhood developmental health. However, their effects on the childhood gut microbiome, which also impacts health, are largely unexplored using microbiome multiomics including the metagenome and metatranscriptome. This study examined the associations of fecal profiles of metal/element exposures with gut microbiome species and active functional pathways in 8- to 12-year-old children (N = 116) participating in the GESTation and Environment (GESTE) cohort study. We analyzed 19 stool metal and element concentrations (B, Na, Mg, Al, K, Ca, V, Cr, Mn, Fe, Co, Ni, Cu, Zn, As, Mo, Cd, Ba, and Pb). Covariate-adjusted linear regression models identified several significant microbiome associations with continuous stool metal/element concentrations. For instance, Zn was positively associated with Turicibacter sanguinis (coef = 1.354, q-value = 0.039) and negatively associated with Eubacterium eligens (coef = -0.794, q-value = 0.044). Higher concentrations of Cd were associated with lower Eubacterium eligens (coef = -0.774, q-value = 0.045). Additionally, a total of 490 significant functional pathways such as biosynthesis and degradation/utilization/assimilation were identified, corresponding to different functions, including amino acid synthesis and carbohydrate degradation. Our results suggest links among metal exposures, pediatric gut microbiome multiomics, and potential health implications. Future work will further explore their relation to childhood health.
Collapse
Affiliation(s)
- Feng Gao
- Department of Environmental Health Sciences, Fielding School of Public Health, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California 90095, United States
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Yike Shen
- Department of Earth and Environmental Sciences, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Hannah E Laue
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst School of Public Health and Health Sciences, Amherst, Massachusetts 01003, United States
| | - Fion K Lau
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Virginie Gillet
- Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Yunjia Lai
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Martha J Shrubsole
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Diddier Prada
- Institute for Health Equity Research - IHER, Department of Population Health Science and Policy and the Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Zhonghua Liu
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York 10032, United States
| | | | - Larissa Takser
- Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Andrea A Baccarelli
- Office of the Dean, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| |
Collapse
|
27
|
Holani R, Bar-Yoseph H, Krekhno Z, Serapio-Palacios A, Moon KM, Stacey RG, Donald KA, Deng W, Bressler B, Magaña AA, Foster LJ, Atser MG, Johnson JD, Finlay B. Bile acid-induced metabolic changes in the colon promote Enterobacteriaceae expansion and associate with dysbiosis in Crohn's disease. Sci Signal 2024; 17:eadl1786. [PMID: 39689182 DOI: 10.1126/scisignal.adl1786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/10/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024]
Abstract
Bile acids (BAs) affect the growth of potentially pathogenic commensals, including those from the Enterobacteriaceae family, which are frequently overrepresented in inflammatory bowel disease (IBD). BAs are normally reabsorbed in the ileum for recycling and are often increased in the colonic lumina of patients with IBD, including those with Crohn's disease (CD). Here, we investigated the influence of BAs on gut colonization by Enterobacteriaceae. We found increased abundance of Enterobacteriaceae in the colonic mucosae of patients with CD with a concomitant decrease in the transporters that resorb BAs in the ileum. The increase in Enterobacteriaceae colonization was greater in the colons of patients who had undergone terminal ileum resection compared with those with intact ileum, leading us to hypothesize that BAs promote intestinal colonization by Enterobacteriaceae. Exposure of human colonic epithelial cell lines to BAs reduced mitochondrial respiration, increased oxygen availability, and enhanced the epithelial adherence of several Enterobacteriaceae members. In a publicly available human dataset, mucosal Enterobacteriaceae was negatively associated with the expression of genes related to mitochondrial function. In a murine model, increased intestinal BA availability enhanced colonization by Escherichia coli in a manner that depended on bacterial respiration. Together, our findings demonstrate that BAs reduce mitochondrial respiration in the colon, leading to an increase in oxygen availability that facilitates Enterobacteriaceae colonization. This identification of BAs as facilitators of host-commensal interactions may be relevant to multiple intestinal diseases.
Collapse
Affiliation(s)
- Ravi Holani
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Haggai Bar-Yoseph
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Zakhar Krekhno
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Antonio Serapio-Palacios
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kyung-Mee Moon
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - Richard G Stacey
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katherine A Donald
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wanyin Deng
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian Bressler
- Division of Gastroenterology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Armando A Magaña
- Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael G Atser
- Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - James D Johnson
- Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Barton Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
28
|
Vaher K, Cabez MB, Parga PL, Binkowska J, van Beveren GJ, Odendaal ML, Sullivan G, Stoye DQ, Corrigan A, Quigley AJ, Thrippleton MJ, Bastin ME, Bogaert D, Boardman JP. The neonatal gut microbiota: A role in the encephalopathy of prematurity. Cell Rep Med 2024; 5:101845. [PMID: 39637857 DOI: 10.1016/j.xcrm.2024.101845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/11/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
Preterm birth correlates with brain dysmaturation and neurocognitive impairment. The gut microbiome associates with behavioral outcomes in typical development, but its relationship with neurodevelopment in preterm infants is unknown. We characterize fecal microbiome in a cohort of 147 neonates enriched for very preterm birth using 16S-based and shotgun metagenomic sequencing. Delivery mode strongly correlates with the preterm microbiome shortly after birth. Low birth gestational age, infant sex assigned at birth, and antibiotics associate with microbiome composition at neonatal intensive care unit discharge. We integrate these data with term-equivalent structural and diffusion brain MRI. Bacterial community composition associates with MRI features of encephalopathy of prematurity. Particularly, abundances of Escherichia coli and Klebsiella spp. correlate with microstructural parameters in deep and cortical gray matter. Metagenome functional capacity analyses suggest that these bacteria may interact with brain microstructure via tryptophan and propionate metabolism. This study indicates that the gut microbiome associates with brain development following preterm birth.
Collapse
Affiliation(s)
- Kadi Vaher
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Manuel Blesa Cabez
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Paula Lusarreta Parga
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Justyna Binkowska
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Gina J van Beveren
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital and University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Mari-Lee Odendaal
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven 3721 MA, the Netherlands
| | - Gemma Sullivan
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - David Q Stoye
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Amy Corrigan
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Alan J Quigley
- Department of Paediatric Radiology, Royal Hospital for Children and Young People, Edinburgh EH16 4TJ, UK
| | | | - Mark E Bastin
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Debby Bogaert
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital and University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - James P Boardman
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
29
|
Huang J, Lu Y, Tian F, Ni Y. Association of body index with fecal microbiome in children cohorts with ethnic-geographic factor interaction: accurately using a Bayesian zero-inflated negative binomial regression model. mSystems 2024; 9:e0134524. [PMID: 39570024 DOI: 10.1128/msystems.01345-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024] Open
Abstract
The exponential growth of high-throughput sequencing (HTS) data on the microbial communities presents researchers with an unparalleled opportunity to delve deeper into the association of microorganisms with host phenotype. However, this growth also poses a challenge, as microbial data are complex, sparse, discrete, and prone to zero inflation. Herein, by utilizing 10 distinct counting models for analyzing simulated data, we proposed an innovative Bayesian zero-inflated negative binomial (ZINB) regression model that is capable of identifying differentially abundant taxa associated with distinctive host phenotypes and quantifying the effects of covariates on these taxa. Our proposed model exhibits excellent accuracy compared with conventional Hurdle and INLA models, especially in scenarios characterized by inflation and overdispersion. Moreover, we confirm that dispersion parameters significantly affect the accuracy of model results, with defects gradually alleviating as the number of analyzed samples increases. Subsequently applying our model to amplicon data in real multi-ethnic children cohort, we found that only a subset of taxa were identified as having zero inflation in real data, suggesting that the prevailing understanding and processing of microbial count data in most previous microbiome studies were overly dogmatic. In practice, our pipeline of integrating bacterial differential abundance in microbiome data and relevant covariates is effective and feasible. Taken together, our method is expected to be extended to the microbiota studies of various multi-cohort populations. IMPORTANCE The microbiome is closely associated with physical indicators of the body, such as height, weight, age and BMI, which can be used as measures of human health. Accurately identifying which taxa in the microbiome are closely related to indicators of physical development is valuable as microbial markers of regional child growth trajectory. Zero-inflated negative binomial (ZINB) model, a type of Bayesian generalized linear model, can be effectively modeled in complex biological systems. We present an innovative ZINB regression model that is capable of identifying differentially abundant taxa associated with distinctive host phenotypes and quantifying the effects of covariates on these taxa, and demonstrate that its accuracy is superior to traditional Hurdle and INLA models. Our pipeline of integrating bacterial differential abundance in microbiome data and relevant covariates is effective and feasible.
Collapse
Affiliation(s)
- Jian Huang
- School of Food Science and Technology, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Special Probiotics and Dairy Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Yanzhuan Lu
- School of Food Science and Technology, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Special Probiotics and Dairy Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yongqing Ni
- School of Food Science and Technology, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Special Probiotics and Dairy Technology, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
30
|
Holman DB, Gzyl KE, Kommadath A. Florfenicol administration in piglets co-selects for multiple antimicrobial resistance genes. mSystems 2024; 9:e0125024. [PMID: 39584815 DOI: 10.1128/msystems.01250-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024] Open
Abstract
Antimicrobial use in food-producing animals such as pigs is a significant issue due to its association with antimicrobial resistance. Florfenicol is a broad-spectrum phenicol antibiotic used in swine for various indications; however, its effect on the swine microbiome and resistome is largely unknown. This study investigated these effects in piglets treated intramuscularly with florfenicol at 1 and 7 days of age. Fecal samples were collected from treated (n = 30) and untreated (n = 30) pigs at nine different time points up until 140 days of age, and the fecal metagenomes were sequenced. The fecal microbiomes of the two groups of piglets were most dissimilar in the immediate period following florfenicol administration. These differences were driven in part by an increase in the relative abundance of Clostridium scindens, Enterococcus faecalis, and Escherichia spp. in the florfenicol-treated piglets and Fusobacterium spp., Pauljensenia hyovaginalis, and Ruminococcus gnavus in the control piglets. In addition to selecting for florfenicol resistance genes (floR, fexA, and fexB), florfenicol also selected for genes conferring resistance to the aminoglycosides, beta-lactams, or sulfonamides up until weaning at 21 days of age. Florfenicol-resistant Escherichia coli isolated from these piglets were found to carry a plasmid with floR, along with tet(A), aph(6)-Id, aph(3″)-Ib, sul2, and blaTEM-1/blaCMY-2. A plasmid carrying fexB and poxtA (phenicols and oxazolidinones) was identified in florfenicol-resistant Enterococcus avium, Enterococcus faecium, and E. faecalis isolates from the treated piglets. This study highlights the potential for co-selection and perturbation of the fecal microbial community in pre-weaned piglets administered florfenicol.IMPORTANCEAntimicrobial use remains a serious challenge in food-animal production due to its linkage with antimicrobial resistance. Antimicrobial resistance can reduce the efficacy of veterinary treatment and can potentially be transferred to humans through the food chain or direct contact with animals and their environment. In this study, early-life florfenicol treatment in piglets altered the composition of the fecal microbiome and selected for many unrelated antimicrobial resistance genes up until weaning at 21 days of age. Part of this co-selection process appeared to involve an Escherichia coli plasmid carrying a florfenicol resistance gene along with genes conferring resistance to at least four other antimicrobial classes. In addition, florfenicol selected for certain genes that provide resistance to multiple antimicrobial classes, including the oxazolidinones. These results highlight that florfenicol can co-select for multiple antimicrobial resistance genes, and their presence on mobile genetic elements suggests the potential for transfer to other bacteria.
Collapse
Affiliation(s)
- Devin B Holman
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, Alberta, Canada
| | - Katherine E Gzyl
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, Alberta, Canada
| | - Arun Kommadath
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, Alberta, Canada
| |
Collapse
|
31
|
Smith KM, Francisco SG, Zhu Y, LeRoith T, Davis ML, Crott JW, Barger K, Greenberg AS, Smith DE, Taylor A, Yeruva L, Rowan S. Dietary prevention of antibiotic-induced dysbiosis and mortality upon aging in mice. FASEB J 2024; 38:e70241. [PMID: 39655692 PMCID: PMC11629448 DOI: 10.1096/fj.202402262r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/08/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024]
Abstract
Oral antibiotic use is both widespread and frequent in older adults and has been linked to dysbiosis of the gut microbiota, enteric infection, and chronic diseases. Diet and nutrients, particularly prebiotics, may modify the susceptibility of the gut microbiome to antibiotic-induced dysbiosis. We fed 12-month-old mice a high glycemic (HG) or low glycemic (LG) diet with or without antibiotics (ampicillin and neomycin) for an additional 11 months. The glycemic index was modulated by the ratio of rapidly digested amylopectin starch to slowly digested amylose, a type-2-resistant starch. We observed a significant decrease in survival of mice fed a HG diet containing antibiotics (HGAbx) relative to those fed a LG diet containing antibiotics (LGAbx). HGAbx mice died with an enlarged and hemorrhagic cecum, which is associated with colonic hyperplasia and goblet cell depletion. Gut microbiome analysis revealed a pronounced expansion of Proteobacteria and a near-complete loss of Bacteroidota and Firmicutes commensal bacteria in HGAbx, whereas the LGAbx group maintained a population of Bacteroides and more closely resembled the LG microbiome. The predicted functional capacity for bile salt hydrolase activity was lost in HGAbx mice but retained in LGAbx mice. An LG diet containing amylose may therefore be a potential therapeutic to prevent antibiotic-induced dysbiosis and morbidity.
Collapse
Affiliation(s)
- Kelsey M. Smith
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| | - Sarah G. Francisco
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Ying Zhu
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| | - Tanya LeRoith
- Department of Biomedical Sciences and PathobiologyVA‐MD College of Veterinary Medicine, Virginia TechBlacksburgVirginiaUSA
| | - Meredith L. Davis
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Jimmy W. Crott
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- Department of Pathology & Laboratory MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Kathryn Barger
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Andrew S. Greenberg
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| | - Donald E. Smith
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Allen Taylor
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| | - Laxmi Yeruva
- USDA‐ARS, Microbiome and Metabolism Research UnitArkansas Children's Nutrition CenterLittle RockArkansasUSA
| | - Sheldon Rowan
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| |
Collapse
|
32
|
Malcomson FC, Louca P, Nelson A, Willis ND, McCallum I, Xie L, Ouwehand AC, Stowell JD, Preston T, Morrison DJ, Kelly SB, Bradburn DM, Belshaw NJ, Johnson IT, Corfe BM, Stewart CJ, Mathers JC. Effects of non-digestible carbohydrates on gut microbiota and microbial metabolites: a randomised, controlled dietary intervention in healthy individuals. Br J Nutr 2024; 132:1433-1445. [PMID: 39494600 PMCID: PMC7616798 DOI: 10.1017/s000711452400271x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
The gut microbiome is impacted by certain types of dietary fibre. However, the type, duration and dose needed to elicit gut microbial changes and whether these changes also influence microbial metabolites remain unclear. This study investigated the effects of supplementing healthy participants with two types of non-digestible carbohydrates (resistant starch (RS) and polydextrose (PD)) on the stool microbiota and microbial metabolite concentrations in plasma, stool and urine, as secondary outcomes in the Dietary Intervention Stem Cells and Colorectal Cancer (DISC) Study. The DISC study was a double-blind, randomised controlled trial that supplemented healthy participants with RS and/or PD or placebo for 50 d in a 2 × 2 factorial design. DNA was extracted from stool samples collected pre- and post-intervention, and V4 16S rRNA gene sequencing was used to profile the gut microbiota. Metabolite concentrations were measured in stool, plasma and urine by high-performance liquid chromatography. A total of fifty-eight participants with paired samples available were included. After 50 d, no effects of RS or PD were detected on composition of the gut microbiota diversity (alpha- and beta-diversity), on genus relative abundance or on metabolite concentrations. However, Drichlet's multinomial mixture clustering-based approach suggests that some participants changed microbial enterotype post-intervention. The gut microbiota and fecal, plasma and urinary microbial metabolites were stable in response to a 50-d fibre intervention in middle-aged adults. Larger and longer studies, including those which explore the effects of specific fibre sub-types, may be required to determine the relationships between fibre intake, the gut microbiome and host health.
Collapse
Affiliation(s)
- Fiona C. Malcomson
- Human Nutrition and Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Centre for Cancer, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Panayiotis Louca
- Human Nutrition and Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Andrew Nelson
- Department of Applied Science, Northumbria University, Newcastle upon Tyne, NE1 8ST, UK
| | - Naomi D. Willis
- Human Nutrition and Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Iain McCallum
- Northumbria Healthcare NHS Foundation Trust, North Tyneside General Hospital, Rake Lane, North Shields, NE29 8NH, UK
| | - Long Xie
- Human Nutrition and Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | | | | | - Tom Preston
- Scottish Universities Environmental Research Centre, College of Science and Engineering, University of Glasgow, Glasgow, UK
| | - Douglas J. Morrison
- Scottish Universities Environmental Research Centre, College of Science and Engineering, University of Glasgow, Glasgow, UK
| | - Seamus B. Kelly
- Northumbria Healthcare NHS Foundation Trust, North Tyneside General Hospital, Rake Lane, North Shields, NE29 8NH, UK
| | - D. Michael Bradburn
- Northumbria Healthcare National Health Service Foundation Trust, Ashington, UK
| | - Nigel J. Belshaw
- University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Ian T. Johnson
- Quadram Institute, Norwich Research Park, Norwich, Norfolk, NR4 7UQ, UK
| | - Bernard M. Corfe
- Human Nutrition and Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Christopher J. Stewart
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - John C. Mathers
- Human Nutrition and Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
33
|
Nickols WA, Kuntz T, Shen J, Maharjan S, Mallick H, Franzosa EA, Thompson KN, Nearing JT, Huttenhower C. MaAsLin 3: Refining and extending generalized multivariable linear models for meta-omic association discovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628459. [PMID: 39713460 PMCID: PMC11661281 DOI: 10.1101/2024.12.13.628459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
A key question in microbial community analysis is determining which microbial features are associated with community properties such as environmental or health phenotypes. This statistical task is impeded by characteristics of typical microbial community profiling technologies, including sparsity (which can be either technical or biological) and the compositionality imposed by most nucleotide sequencing approaches. Many models have been proposed that focus on how the relative abundance of a feature (e.g. taxon or pathway) relates to one or more covariates. Few of these, however, simultaneously control false discovery rates, achieve reasonable power, incorporate complex modeling terms such as random effects, and also permit assessment of prevalence (presence/absence) associations and absolute abundance associations (when appropriate measurements are available, e.g. qPCR or spike-ins). Here, we introduce MaAsLin 3 (Microbiome Multivariable Associations with Linear Models), a modeling framework that simultaneously identifies both abundance and prevalence relationships in microbiome studies with modern, potentially complex designs. MaAsLin 3 also newly accounts for compositionality with experimental (spike-ins and total microbial load estimation) or computational techniques, and it expands the space of biological hypotheses that can be tested with inference for new covariate types. On a variety of synthetic and real datasets, MaAsLin 3 outperformed current state-of-the-art differential abundance methods in testing and inferring associations from compositional data. When applied to the Inflammatory Bowel Disease Multi-omics Database, MaAsLin 3 corroborated many previously reported microbial associations with the inflammatory bowel diseases, but notably 77% of associations were with feature prevalence rather than abundance. In summary, MaAsLin 3 enables researchers to identify microbiome associations with higher accuracy and more specific association types, especially in complex datasets with multiple covariates and repeated measures.
Collapse
Affiliation(s)
- William A. Nickols
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Thomas Kuntz
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Jiaxian Shen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sagun Maharjan
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Himel Mallick
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Statistics and Data Science, Cornell University, Ithaca, NY
| | - Eric A. Franzosa
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kelsey N. Thompson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jacob T. Nearing
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| |
Collapse
|
34
|
Aluja M, Cerqueda-García D, Altúzar-Molina A, Guillén L, Acosta-Velasco E, Conde-Alarcón J, Moya A. Geographic variation and core microbiota composition of Anastrepha ludens (Diptera: Tephritidae) infesting a single host across latitudinal and altitudinal gradients. PeerJ 2024; 12:e18555. [PMID: 39686986 PMCID: PMC11648694 DOI: 10.7717/peerj.18555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/29/2024] [Indexed: 12/18/2024] Open
Abstract
Anastrepha ludens is a pestiferous tephritid fly species exhibiting extreme polyphagy. It develops optimally in hosts rich in sugar but low nitrogen content. We studied the geographical influence on the composition of A. ludens's larval and newly emerged adult gut microbiota in altitudinal (0-2,000 masl) and latitudinal (ca. 800 km from 17° to 22°N latitude) transects along the coastline of the state of Veracruz, Mexico. In the 16 collection sites, we only collected Citrus x aurantium fruit (238 samples of A. ludens larvae and adults, plus 73 samples of pulp) to control for host effect, hypothesizing that there exists a conserved core microbiota that would be dominated by nitrogen-fixing bacteria. We found that latitude triggered more significant changes in the gut microbiota than altitude. Northern and southernmost samples differed the most in microbiota composition, with a trade-off between Acetobacteraceae and Rhizobiaceae driving these differences. As hypothesized, the core microbiota in each sampling site, contained the functional group of nitrogen-fixing bacteria. We conclude that A. ludens larvae can acquire multiple diazotrophic symbionts along its wide distribution range where it infests fruit with a high C:N ratio in the pulp.
Collapse
Affiliation(s)
- Martín Aluja
- Red de Manejo Biorracional de Plagas y Vectores, Clúster Científico y Tecnológico BioMimic, Instituto de Ecología, A.C.–INECOL, El Haya, Xalapa, Veracruz, Mexico
| | - Daniel Cerqueda-García
- Red de Manejo Biorracional de Plagas y Vectores, Clúster Científico y Tecnológico BioMimic, Instituto de Ecología, A.C.–INECOL, El Haya, Xalapa, Veracruz, Mexico
| | - Alma Altúzar-Molina
- Red de Manejo Biorracional de Plagas y Vectores, Clúster Científico y Tecnológico BioMimic, Instituto de Ecología, A.C.–INECOL, El Haya, Xalapa, Veracruz, Mexico
| | - Larissa Guillén
- Red de Manejo Biorracional de Plagas y Vectores, Clúster Científico y Tecnológico BioMimic, Instituto de Ecología, A.C.–INECOL, El Haya, Xalapa, Veracruz, Mexico
| | - Emilio Acosta-Velasco
- Red de Manejo Biorracional de Plagas y Vectores, Clúster Científico y Tecnológico BioMimic, Instituto de Ecología, A.C.–INECOL, El Haya, Xalapa, Veracruz, Mexico
| | - Juan Conde-Alarcón
- Red de Manejo Biorracional de Plagas y Vectores, Clúster Científico y Tecnológico BioMimic, Instituto de Ecología, A.C.–INECOL, El Haya, Xalapa, Veracruz, Mexico
| | - Andrés Moya
- Instituto de Biología Integrativa de Sistemas (I2SysBio), Universidad de Valencia and Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| |
Collapse
|
35
|
Peng Y, Zhu J, Wang S, Liu Y, Liu X, DeLeon O, Zhu W, Xu Z, Zhang X, Zhao S, Liang S, Li H, Ho B, Ching JYL, Cheung CP, Leung TF, Tam WH, Leung TY, Chang EB, Chan FKL, Zhang L, Ng SC, Tun HM. A metagenome-assembled genome inventory for children reveals early-life gut bacteriome and virome dynamics. Cell Host Microbe 2024; 32:2212-2230.e8. [PMID: 39591974 DOI: 10.1016/j.chom.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/03/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024]
Abstract
Existing microbiota databases are biased toward adult samples, hampering accurate profiling of the infant gut microbiome. Here, we generated a metagenome-assembled genome inventory for children (MAGIC) from a large collection of bulk and viral-like particle-enriched metagenomes from 0 to 7 years of age, encompassing 3,299 prokaryotic and 139,624 viral species-level genomes, 8.5% and 63.9% of which are unique to MAGIC. MAGIC improves early-life microbiome profiling, with the greatest improvement in read mapping observed in Africans. We then identified 54 candidate keystone species, including several Bifidobacterium spp. and four phages, forming guilds that fluctuated in abundance with time. Their abundances were reduced in preterm infants and were associated with childhood allergies. By analyzing the B. longum pangenome, we found evidence of phage-mediated evolution and quorum sensing-related ecological adaptation. Together, the MAGIC database recovers genomes that enable characterization of the dynamics of early-life microbiomes, identification of candidate keystone species, and strain-level study of target species.
Collapse
Affiliation(s)
- Ye Peng
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Jie Zhu
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Shilan Wang
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Yingzhi Liu
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Xin Liu
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Orlando DeLeon
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, The University of Chicago, Chicago, IL 60637, USA
| | - Wenyi Zhu
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Zhilu Xu
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Xi Zhang
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Shilin Zhao
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Suisha Liang
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China
| | - Hang Li
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China
| | - Brian Ho
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China
| | - Jessica Yuet-Ling Ching
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Chun Pan Cheung
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Ting Fan Leung
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Wing Hung Tam
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Tak Yeung Leung
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Eugene B Chang
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, The University of Chicago, Chicago, IL 60637, USA
| | - Francis Ka Leung Chan
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Lin Zhang
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China.
| | - Siew Chien Ng
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR 999077, China.
| | - Hein Min Tun
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China; Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China.
| |
Collapse
|
36
|
Da-Anoy J, Posadas N, Conaco C. Interspecies differences in the transcriptome response of corals to acute heat stress. PeerJ 2024; 12:e18627. [PMID: 39677947 PMCID: PMC11639872 DOI: 10.7717/peerj.18627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024] Open
Abstract
Rising sea surface temperatures threaten the survival of corals worldwide, with coral bleaching events becoming more commonplace. However, different coral species are known to exhibit variable levels of susceptibility to thermal stress. To elucidate genetic mechanisms that may underlie these differences, we compared the gene repertoire of four coral species, Favites colemani, Montipora digitata, Acropora digitifera, and Seriatopora caliendrum, that were previously demonstrated to have differing responses to acute thermal stress. We found that more tolerant species, like F. colemani and M. digitata, possess a greater abundance of antioxidant protein families and chaperones. Under acute thermal stress conditions, only S. caliendrum showed a significant bleaching response, which was accompanied by activation of the DNA damage response network and drastic upregulation of stress response genes (SRGs). This suggests that differences in SRG orthologs, as well as the mechanisms that control SRG expression response, contribute to the ability of corals to maintain stability of physiological functions required to survive shifts in seawater temperature.
Collapse
Affiliation(s)
- Jeric Da-Anoy
- Marine Science Institute, University of the Philippines Diliman, Quezon City, Philippines
- Department of Biology, Boston University, Boston, MA, United States of America
| | - Niño Posadas
- Marine Science Institute, University of the Philippines Diliman, Quezon City, Philippines
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Cecilia Conaco
- Marine Science Institute, University of the Philippines Diliman, Quezon City, Philippines
| |
Collapse
|
37
|
Zamudio-Flores J, Cerqueda D, Phillips-Farfán B, Guerrero-Flores S, Salinas-García AF, Meléndez-Herrera E, Sélem-Mojica N, Kline AE, Lajud N. Environmental enrichment-induced cognitive recovery after a moderate pediatric traumatic brain injury is associated with the gut microbiota and neuroinflammation. Exp Neurol 2024; 385:115109. [PMID: 39662794 DOI: 10.1016/j.expneurol.2024.115109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/11/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Pediatric traumatic brain injury (TBI) is a significant health concern, yet access to rehabilitation therapies for children remains limited. Environmental enrichment (EE) is a preclinical model of neurorehabilitation that promotes behavioral recovery and reduces neuroinflammation after TBI. While the gut microbiota has recently emerged as a potential therapeutic target for treating TBI sequelae in adults, its role in recovery after pediatric TBI remains unclear. Therefore, our aim was to assess the effect of EE on gut microbiota and its correlation with cognition as well as microglial morphology in a preclinical model of pediatric TBI. Male rats underwent a controlled cortical impact of moderate severity or sham injury at postnatal day 21 and were then randomly assigned to either EE or standard (STD) housing. Cognition was evaluated using the Morris water maze (MWM) on post-injury days 14-19. Microglial morphology and caecum microbiota was characterized on post-injury day 21. Cognitive deficits and increased microglial activation in the ipsilateral cortex were observed in the STD-housed TBI rats but not those in EE. TBI decreased microbiota α-diversity, while PERMANOVA analysis showed that both TBI and EE modified microbiota β-diversity. Furthermore, regression models indicated that microglial morphology in the ipsilateral cortex and Lactobacillus reuteri predicted behavioral outcomes, while Prevotellaceae NK3B31 was associated with microglial morphology. The data suggest that EE mitigates TBI-induced alterations in gut microbiota and that there is a complex interplay between EE, microbiota and microglial morphology that predicts behavioral recovery in pediatric rats.
Collapse
Affiliation(s)
- Jonathan Zamudio-Flores
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico; Instituto de Investigaciones sobre los Recursos Naturales - Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Daniel Cerqueda
- Red de Manejo Biorracional de Plagas y Vectores, Instituto de Ecología, A. C., Xalapa, Mexico
| | | | | | - Ana Fernanda Salinas-García
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico; Instituto de Investigaciones sobre los Recursos Naturales - Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Esperanza Meléndez-Herrera
- Instituto de Investigaciones sobre los Recursos Naturales - Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Nelly Sélem-Mojica
- Centro de Ciencias Matemáticas, Universidad Nacional Autónoma de, Mexico
| | - Anthony E Kline
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States of America; Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; Psychology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Naima Lajud
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico.
| |
Collapse
|
38
|
Cui Z, Wang S, Niu J, Ma J, Yang H. Bifidobacterium species serve as key gut microbiome regulators after intervention in gestational diabetes mellitus. BMC Microbiol 2024; 24:520. [PMID: 39643877 PMCID: PMC11622461 DOI: 10.1186/s12866-024-03680-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024] Open
Abstract
Gut microbiome dysbiosis is associated with gestational diabetes mellitus (GDM), and its modulation represents a promising approach for enhancing glycemic control. In this study, we aimed to discover specific alterations in the gut microbiome through lifestyle management. We performed metagenome sequencing on fecal samples and measured short-chain fatty acid (SCFA) in plasma samples from 27 well-controlled GDM pregnancies before and after glycemic control. At the same time, 38 normal glucose tolerance (NGT) samples served as controls. Additionally, we employed two-sample Mendelian Randomization (MR) to validate our findings against Genome-Wide Association Study (GWAS) database. Our dynamic analysis revealed Bifidobacterium genus increased in GDM patients after intervention. The MR analysis confirmed that the family of Bifidobacteriaceae (OR 0.929, 95% CI, 0.886-0.975; P = 0.003) was the only negatively associated family with GDM. Further analysis indicated the increased abundance of Bifidobacterium species were negatively correlated with glycemic traits (Spearman rho mean - 0.32 ± 0.34) but positively correlated with plasma SCFA levels (Spearman rho mean 0.24 ± 0.19). Functional analysis revealed that the quorum-sensing pathway had the strongest effect on the ability of Bifidobacterium to promote glucose homeostasis (Spearman rho = -0.34), suggesting its role in regulating intestinal microbiota. Finally, the multivariable MR analysis demonstrated that two pathways, COLANSYN PWY and PWY 7323, responsible for cell surface compound synthesis in gram-negative bacteria, mediated 14.83% (P = 0.017) and 16.64% (P = 0.049) of the protective effects of Bifidobacteriaceae against GDM, respectively. In summary, Bifidobacterium is an effective gut microbiota regulator for GDM-related glucose homeostasis.
Collapse
Affiliation(s)
- Zifeng Cui
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Shuxian Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Jianhua Niu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Jingmei Ma
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China.
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China.
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China.
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China.
| |
Collapse
|
39
|
Davies JM, Teh JJ, Ewais T, Begun J. Does Improving Depression Symptoms in Young Adults With Inflammatory Bowel Disease Alter Their Microbiome? Inflamm Bowel Dis 2024; 30:2428-2439. [PMID: 38839073 PMCID: PMC11630472 DOI: 10.1093/ibd/izae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Indexed: 06/07/2024]
Abstract
BACKGROUND Patients with inflammatory bowel diseases (IBDs) are more likely to have depression and anxiety symptoms compared with healthy individuals and those with other chronic illnesses. Previous studies have shown a link between the microbiome composition and depression symptoms; however, many antidepressant medications have antibacterial activity confounding cross-sectional studies of these populations. Therefore, we aimed to determine whether we could detect longitudinal changes in the microbiome of a subset of patients who participated in a previously published mindfulness-based cognitive therapy (MBCT) study to improve depression symptoms in adolescents and young adults with IBD. METHODS Stool samples were collected at baseline and 8 weeks (n = 24 participants, 37 total samples, 13 paired samples). During this time, some participants achieved a 50% reduction in their depression symptoms either through MBCT or treatment as usual with their mental health team (responders). The microbiome composition and function of responders were compared with participants who did not improve their depression scores (nonresponders). Depression scores were determined using the depression, anxiety, and stress score (DASS-21), and metagenomic sequencing of stool samples was performed. RESULTS No difference in alpha diversity was found between responders and nonresponders. Beta diversity measures were similarly unchanged. Clinical features including fecal calprotectin, C-reactive protein, and serum IL-6 levels were unchanged. CONCLUSIONS In this small longitudinal study, we were not able to detect longitudinal changes in the microbiome associated with improvement in depression scores. Follow-up studies that are sufficiently powered to detect changes in the microbiome are required to confirm our results.
Collapse
Affiliation(s)
- Julie M Davies
- Mater Research-The University of Queensland, Woolloongabba, QLD, Australia
| | - Jing Jie Teh
- Frazer Institute, The University of Queensland, Woolloongabba QLD, Australia
| | - Tatjana Ewais
- Mater Adolescent and Young Adult Health Clinic, South Brisbane, QLD, Australia
- School of Medicine, The University of Queensland, St Lucia, QLD, Australia
- School of Medicine and Dentistry, Gold Coast Campus, Griffith University, Southport, QLD, Australia
| | - Jakob Begun
- Mater Research-The University of Queensland, Woolloongabba, QLD, Australia
- Department of Gastroenterology, Mater Hospital Brisbane, South Brisbane, Australia
| |
Collapse
|
40
|
Rahal Z, Liu Y, Peng F, Yang S, Jamal MA, Sharma M, Moreno H, Damania AV, Wong MC, Ross MC, Sinjab A, Zhou T, Chen M, Reischle IT, Feng J, Chukwuocha C, Tang E, Abaya C, Lim JK, Leung CH, Lin HY, Deboever N, Lee JJ, Sepesi B, Gibbons DL, Wargo JA, Fujimoto J, Wang L, Petrosino JF, Ajami NJ, Jenq RR, Moghaddam SJ, Cascone T, Hoffman K, Kadara H. Inflammation Mediated by Gut Microbiome Alterations Promotes Lung Cancer Development and an Immunosuppressed Tumor Microenvironment. Cancer Immunol Res 2024; 12:1736-1752. [PMID: 39269772 PMCID: PMC11614694 DOI: 10.1158/2326-6066.cir-24-0469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/17/2024] [Accepted: 09/13/2024] [Indexed: 09/15/2024]
Abstract
Accumulating evidence indicates that the gut microbiome influences cancer progression and therapy. We recently showed that progressive changes in gut microbial diversity and composition are closely coupled with tobacco-associated lung adenocarcinoma in a human-relevant mouse model. Furthermore, we demonstrated that the loss of the antimicrobial protein Lcn2 in these mice exacerbates protumor inflammatory phenotypes while further reducing microbial diversity. Yet, how gut microbiome alterations impinge on lung adenocarcinoma development remains poorly understood. In this study, we investigated the role of gut microbiome changes in lung adenocarcinoma development using fecal microbiota transfer and delineated a pathway by which gut microbiome alterations incurred by loss of Lcn2 fostered the proliferation of proinflammatory bacteria of the genus Alistipes, triggering gut inflammation. This inflammation propagated systemically, exerting immunosuppression within the tumor microenvironment, augmenting tumor growth through an IL6-dependent mechanism and dampening response to immunotherapy. Corroborating our preclinical findings, we found that patients with lung adenocarcinoma with a higher relative abundance of Alistipes species in the gut showed diminished response to neoadjuvant immunotherapy. These insights reveal the role of microbiome-induced inflammation in lung adenocarcinoma and present new potential targets for interception and therapy.
Collapse
Affiliation(s)
- Zahraa Rahal
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuejiang Liu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
| | - Fuduan Peng
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sujuan Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohamed A. Jamal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manvi Sharma
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hannah Moreno
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Ashish V. Damania
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Departments of Platform for Innovative Microbiome and Translational Research (PRIME-TR), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew C. Wong
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Departments of Platform for Innovative Microbiome and Translational Research (PRIME-TR), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew C. Ross
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Ansam Sinjab
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tieling Zhou
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Minyue Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
| | - Inti Tarifa Reischle
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiping Feng
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chidera Chukwuocha
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth Tang
- Department of Physics, University of Illinois Urbana-Champaign, Urbana-Champaign, IL, USA
| | - Camille Abaya
- Department of Biology, Trinity University, San Antonio, TX, USA
| | - Jamie K Lim
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, John Hopkins University, Baltimore, MD, USA
| | - Cheuk Hong Leung
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Y. Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nathaniel Deboever
- Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, TX, USA
| | - Jack J. Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boris Sepesi
- Departments of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L. Gibbons
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer A. Wargo
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Departments of Platform for Innovative Microbiome and Translational Research (PRIME-TR), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Junya Fujimoto
- Clinical Research Center, Hiroshima University Hospital, Hiroshima, Japan
| | - Linghua Wang
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph F. Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Nadim J Ajami
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Departments of Platform for Innovative Microbiome and Translational Research (PRIME-TR), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert R. Jenq
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Seyed Javad Moghaddam
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristi Hoffman
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
| |
Collapse
|
41
|
Drosdowech S, Bezner S, Daisley B, Chiasson M, Easton A, Rooney N, Huyben D. Influence of feeding black soldier fly (Hermetia illucens), cricket (Gryllodes sigillatus), and superworm (Zophobas morio) on the gut microbiota of rainbow trout (Oncorhynchus mykiss). J Appl Microbiol 2024; 135:lxae295. [PMID: 39580363 DOI: 10.1093/jambio/lxae295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/31/2024] [Accepted: 11/22/2024] [Indexed: 11/25/2024]
Abstract
AIM This study investigates how replacing fishmeal and fish oil with insect meals in feed impacts the gut microbiota in rainbow trout (Oncorhynchus mykiss), a crucial species in aquaculture. METHODS AND RESULTS Dietary inclusion of black soldier fly (Hermetia illucens), cricket (Gryllodes sigillatus), and superworm (Zophobas morio) were evaluated for their impact on intestinal microbial diversity and community composition following a 12-week feeding trial. Fish were fed one of four isonitrogenous and isoenergetic diets: a control diet without insect meal, and diets with 15% defatted black soldier fly meal, full-fat adult cricket meal, or full-fat superworm meal. The microbiota of intestinal digesta and fish feed was characterized using 16S rRNA gene sequencing on the Illumina MiSeq platform. Results revealed significantly lower alpha diversity indices in the cricket treatment compared to the control. Beta diversity analysis showed Bacillota as the dominant phylum across all treatments, with the initial stock population richer in Mycoplasmatota. A novel genus within Mycoplasmataceae was prevalent at Day 0 and in all treatments. Black soldier fly meal increased an unidentified Peptostreptococcaceae genus (bsv123) compared to control and superworm diets, while cricket meal elevated Streptococcus levels. CONCLUSIONS Insect-based diets, particularly with black soldier fly meal, significantly alter beta diversity within the gut microbiota of rainbow trout, with cricket meal reducing alpha diversity and superworm having minimal impact.
Collapse
Affiliation(s)
- Sonja Drosdowech
- School of Environmental Sciences, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| | - Samantha Bezner
- Department of Animal Biosciences, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| | - Brendan Daisley
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| | - Marcia Chiasson
- Ontario Aquaculture Research Centre, University of Guelph, 6957 8 Line W, Elora, Ontario N0B 1S0, Canada
| | - Anne Easton
- Ontario Aquaculture Research Centre, University of Guelph, 6957 8 Line W, Elora, Ontario N0B 1S0, Canada
| | - Neil Rooney
- School of Environmental Sciences, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| | - David Huyben
- Department of Animal Biosciences, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
42
|
Sun Z, Song K. GEMimp: An Accurate and Robust Imputation Method for Microbiome Data Using Graph Embedding Neural Network. J Mol Biol 2024; 436:168841. [PMID: 39490678 DOI: 10.1016/j.jmb.2024.168841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Microbiome research has increasingly underscored the profound link between microbial compositions and human health, with numerous studies establishing a strong correlation between microbiome characteristics and various diseases. However, the analysis of microbiome data is frequently compromised by inherent sparsity issues, characterized by a substantial presence of observed zeros. These zeros not only skew the abundance distribution of microbial species but also undermine the reliability of scientific conclusions drawn from such data. Addressing this challenge, we introduce GEMimp, an innovative imputation method designed to infuse robustness into microbiome data analysis. GEMimp leverages the node2vec algorithm, which incorporates both Breadth-First Search (BFS) and Depth-First Search (DFS) strategies in its random walks sampling process. This approach enables GEMimp to learn nuanced, low-dimensional representations of each taxonomic unit, facilitating the reconstruction of their similarity networks with unprecedented accuracy. Our comparative analysis pits GEMimp against state-of-the-art imputation methods including SAVER, MAGIC and mbImpute. The results unequivocally demonstrate that GEMimp outperforms its counterparts by achieving the highest Pearson correlation coefficient when compared to the original raw dataset. Furthermore, GEMimp shows notable proficiency in identifying significant taxa, enhancing the detection of disease-related taxa and effectively mitigating the impact of sparsity on both simulated and real-world datasets, such as those pertaining to Type 2 Diabetes (T2D) and Colorectal Cancer (CRC). These findings collectively highlight the strong effectiveness of GEMimp, allowing for better analysis on microbial data. With alleviation of sparsity issues, it could be greatly facilitated in downstream analyses and even in the field of microbiology.
Collapse
Affiliation(s)
- Ziwei Sun
- School of Mathematics and Statistics, Qingdao University, Qingdao, China.
| | - Kai Song
- School of Mathematics and Statistics, Qingdao University, Qingdao, China.
| |
Collapse
|
43
|
Mizuno H, Kawamoto S, Uemura K, Park JH, Hori N, Okumura Y, Konishi Y, Hara E. B cell senescence promotes age-related changes in oral microbiota. Aging Cell 2024; 23:e14304. [PMID: 39123277 PMCID: PMC11634744 DOI: 10.1111/acel.14304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/27/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
In recent years, there has been increasing attention towards understanding the relationship between age-related alterations in the oral microbiota and age-associated diseases, with reports emphasizing the significance of maintaining a balanced oral microbiota for host health. However, the precise mechanisms underlying age-related changes in the oral microbiota remain elusive. We recently reported that cellular senescence of ileal germinal center (GC) B cells, triggered by the persistent presence of commensal bacteria, results in diminished IgA production with aging and subsequent alterations in the gut microbiota. Consequently, we hypothesize that a similar phenomenon may occur in the oral cavity, potentially contributing to age-related changes in the oral microbiota. Examination of p16-luc mice, wherein the expression of the senescent cell marker p16INK4a can be visualized, raised under specific pathogen-free (SPF) or germ-free (GF) conditions, indicated that, unlike ileal GC B cells, the accumulation of senescent cells in GC B cells of cervical lymph nodes increases with age regardless of the presence of commensal bacteria. Furthermore, longitudinal studies utilizing the same individual mice throughout their lifespan revealed concurrent age-related alterations in the composition of the oral microbiota and a decline in salivary IgA secretion. Further investigation involving Rag1-/- mice transplanted with B cells from wild-type or p16INK4a and p21Waf1/Cip1 -double knockout mice unveiled that B cell senescence leads to reduced IgA secretion and alteration of the oral microbiota. These findings advance our understanding of the mechanism of age-associated changes in the oral microbiota and open up possibilities of their control.
Collapse
Affiliation(s)
- Hiroya Mizuno
- Department of Molecular Biology, Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Shimpei Kawamoto
- Department of Molecular Biology, Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Ken Uemura
- Department of Molecular Biology, Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Jeong Hoon Park
- Department of Molecular Biology, Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Nozomi Hori
- Department of Molecular Biology, Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Yumiko Okumura
- Department of Molecular Biology, Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Yusuke Konishi
- Department of Molecular Biology, Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Eiji Hara
- Department of Molecular Biology, Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
- Laboratory of Aging Biology, Immunology Frontier Research CenterOsaka UniversitySuitaOsakaJapan
- Center for Infectious Disease Education and ResearchOsaka UniversitySuitaOsakaJapan
| |
Collapse
|
44
|
Mahdavi M, Prévost K, Balthazar P, Hus IFP, Duchesne É, Dumont N, Gagné-Ouellet V, Gagnon C, Laforest-Lapointe I, Massé E. Disturbance of the human gut microbiota in patients with Myotonic Dystrophy type 1. Comput Struct Biotechnol J 2024; 23:2097-2108. [PMID: 38803516 PMCID: PMC11128782 DOI: 10.1016/j.csbj.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a rare autosomal dominant genetic disorder. Although DM1 is primarily characterized by progressive muscular weakness, it exhibits many multisystemic manifestations, such as cognitive deficits, cardiac conduction abnormalities, and cataracts, as well as endocrine and reproductive issues. Additionally, the gastrointestinal (GI) tract is frequently affected, encompassing the entire digestive tract. However, the underlying causes of these GI symptoms remain uncertain, whether it is biomechanical problems of the intestine, involvement of bacterial communities, or both. The primary objective of this study is to investigate the structural changes in the gut microbiome of DM1 patients. To achieve this purpose, 35 patients with DM1 were recruited from the DM-Scope registry of the neuromuscular clinic in the Saguenay-Lac-St-Jean region of the province of Québec, Canada. Stool samples from these 35 patients, including 15 paired samples with family members living with them as controls, were collected. Subsequently, these samples were sequenced by 16S MiSeq and were analyzed with DADA2 to generate taxonomic signatures. Our analysis revealed that the DM1 status correlated with changes in gut bacterial community. Notably, there were differences in the relative abundance of Bacteroidota, Euryarchaeota, Fusobacteriota, and Cyanobacteria Phyla compared to healthy controls. However, no significant shift in gut microbiome community structure was observed between DM1 phenotypes. These findings provide valuable insights into how the gut bacterial community, in conjunction with biomechanical factors, could potentially influence the gastrointestinal tract of DM1 patients.
Collapse
Affiliation(s)
- Manijeh Mahdavi
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, QC J1E 4K8, Canada
| | - Karine Prévost
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, QC J1E 4K8, Canada
| | - Philippe Balthazar
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, QC J1E 4K8, Canada
| | - Isabelle Fisette-Paul Hus
- Department of Rehabilitation, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Élise Duchesne
- Physiotherapy teaching unit, Université du Québec à Chicoutimi, Chicoutimi, G7H 2B1, Canada
| | - Nicolas Dumont
- School of Rehabilitation, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Valérie Gagné-Ouellet
- Department of Rehabilitation, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Cynthia Gagnon
- Department of Rehabilitation, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | | | - Eric Massé
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, QC J1E 4K8, Canada
| |
Collapse
|
45
|
Liu YC, Ramiro-Garcia J, O'Connor S, Paulo LM, Maria Braguglia C, Cristina Gagliano M, O'Flaherty V. Microbial community response to temperature reduction during anaerobic treatment of long chain fatty acids-containing wastewater. BIORESOURCE TECHNOLOGY 2024; 413:131529. [PMID: 39321934 DOI: 10.1016/j.biortech.2024.131529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/29/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
Acclimating mesophilic biomass to low temperatures have been used to start-up psychrophilic anaerobic reactors, but limited microbial information is available during the acclimation. To investigate microbial responses to temperature reductions, duplicate lab-scale anaerobic digestion (AD) reactors were operated for 166 days, with the temperature being reduced from 37°C to 15°C, using synthetic long chain fatty acid (LCFA)-containing wastewater as the feedstock. The acclimated biomass at 15°C exhibited efficient removal of organic matter (total COD>75%, soluble COD>88%, and LCFA>99%). Temperature reductions lead to significant reductions in microbiome diversity. Fermentative bacteria were highly dynamic and functional redundant during temperature reductions. Smithella was the dominant syntrophic bacteria involved in LCFA degradation coupled with Methanothrix and Methanocorpusculum at 15°C. Membrane modifications and compatible cellular solutes production were triggered by temperature reductions as microbial response to cold stress. This study provided molecular insights in microbial acclimation to low temperatures for psychrophilic AD.
Collapse
Affiliation(s)
- Yu-Chen Liu
- Microbial Ecology Laboratory, School of Biological and Chemical Sciences and Ryan Institute, University of Galway, University Road, Galway H91 TK33, Ireland.
| | - Javier Ramiro-Garcia
- Instituto de la Grasa. Consejo Superior de Investigaciones Científicas, Campus Universitario Pablo de Olavide- Ed. 46, Ctra. de Utrera, km. 1, Seville 41013, Spain
| | - Sandra O'Connor
- Microbial Ecology Laboratory, School of Biological and Chemical Sciences and Ryan Institute, University of Galway, University Road, Galway H91 TK33, Ireland
| | - Lara M Paulo
- Microbial Ecology Laboratory, School of Biological and Chemical Sciences and Ryan Institute, University of Galway, University Road, Galway H91 TK33, Ireland
| | - Camilla Maria Braguglia
- Water Research Institute, CNR, Area di Ricera RM1-Montelibretti, Via Salaria km 29.300, 00015 Monterotondo (Roma), Italy
| | - Maria Cristina Gagliano
- Wetsus, European Centre of Excellence for Sustainable Water Technology, Oostergoweg 9, MA 8911 Leeuwarden, the Netherlands
| | - Vincent O'Flaherty
- Microbial Ecology Laboratory, School of Biological and Chemical Sciences and Ryan Institute, University of Galway, University Road, Galway H91 TK33, Ireland
| |
Collapse
|
46
|
Gantt SE, Kemp KM, Colin PL, Hoadley KD, LaJeunesse TC, Warner ME, Kemp DW. Influence of reef habitat on coral microbial associations. ENVIRONMENTAL MICROBIOLOGY REPORTS 2024; 16:e70051. [PMID: 39517101 PMCID: PMC11549029 DOI: 10.1111/1758-2229.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Corals have complex symbiotic associations that can be influenced by the environment. We compare symbiotic dinoflagellate (family: Symbiodiniaceae) associations and the microbiome of five scleractinian coral species from three different reef habitats in Palau, Micronesia. Although pH and temperature corresponded with specific host-Symbiodiniaceae associations common to the nearshore and offshore habitats, bacterial community dissimilarity analyses indicated minimal influence of these factors on microbial community membership for the corals Coelastrea aspera, Psammocora digitata, and Pachyseris rugosa. However, coral colonies sampled close to human development exhibited greater differences in microbial community diversity compared to the nearshore habitat for the coral species Coelastrea aspera, Montipora foliosa, and Pocillopora acuta, and the offshore habitat for Coelastrea aspera, while also showing less consistency in Symbiodiniaceae associations. These findings indicate the influence that habitat location has on the bacterial and Symbiodiniaceae communities comprising the coral holobiont and provide important considerations for the conservation of coral reef communities, especially for island nations with increasing human populations and development.
Collapse
Affiliation(s)
- Shelby E. Gantt
- Department of BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Keri M. Kemp
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | | | - Kenneth D. Hoadley
- Department of Biological SciencesUniversity of AlabamaTuscaloosaAlabamaUSA
- Dauphin Island Sea LabDauphin IslandAlabamaUSA
| | - Todd C. LaJeunesse
- Department of BiologyPennsylvania State UniversityState CollegePennsylvaniaUSA
| | - Mark E. Warner
- School of Marine Science and PolicyUniversity of DelawareLewesDelawareUSA
| | - Dustin W. Kemp
- Department of BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
47
|
Bortoluzzi C, Segura-Wang M, Aureli R, Leduc A, Iuspa MA, Cowieson AJ. Supplementation of precision biotic leads to improved growth performance by modulating the microbiome of broiler chickens fed corn or wheat-based diets. Poult Sci 2024; 103:104451. [PMID: 39504818 PMCID: PMC11577187 DOI: 10.1016/j.psj.2024.104451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
The objective of the present study was to evaluate the effect of a precision biotic (PB; Symphiome®, dsm-firmenich) on the growth performance, nutrient digestibility, litter quality, blood uric acid (UA) concentration, cecal short-chain fatty acids (SCFA) concentration, and ileal and cecal microbiome of broiler chickens fed corn or wheat-based diets. One-day-old broiler chicks were placed in a completely randomized design (2 × 2 factorial scheme) in 4 treatments, 8 replicates/treatment, and 16 birds/replicate. The treatments were a corn or wheat and soybean meal-based diet without or with supplementation of 0.9 kg/MT of PB. All the birds were vaccinated against coccidiosis at the time of placement (Paracox 5). Growth performance was evaluated weekly and at the end of the experiment. At d 35, blood was collected to determine UA concentration and cecal content for SCFA concentration analysis. Ileal digesta was also collected for nutrient digestibility analysis, and ileal digesta and cecal content were collected for microbiome analysis. The data were submitted to two-way ANOVA (P ≤ 0.05), and LSM was used to separate the means in case of interaction. During the starter and grower phases of the study, a better body weight gain (P = 0.0008 and P = 0.04, respectively) was observed in birds fed wheat compared to corn-based diets. From 28 to 36 d of age, the supplementation of PB increased feed intake (P = 0.05), and tended (P = 0.06) to improve the feed conversion ratio by 4.3 points vs non-supplementated birds. Birds fed wheat-based diets had higher (P = 0.02) blood UA than corn based-diets fed birds, and the supplementation of PB led to a reduction (P = 0.02) of blood UA compared to non-supplemented birds. The supplementation of PB changed the abundance of core metabolic pathways of the microbiome, mostly related to protein metabolism, which led to a reduction in blood UA concentration and increase of cecal SCFA concentrations. In conclusion, by beneficially modulating the microbiome, the supplementation of PB was translated into improved growth performance of broiler chickens fed corn or wheat-based diets.
Collapse
Affiliation(s)
| | - M Segura-Wang
- dsm-firmenich, Animal Nutrition and Health R&D Center, Tulln, Austria
| | - R Aureli
- dsm-firmenich, Animal Nutrition and Health R&D Center, Village-Neuf, France
| | - A Leduc
- dsm-firmenich, Animal Nutrition and Health R&D Center, Village-Neuf, France
| | | | | |
Collapse
|
48
|
Oh VKS, Li RW. Wise Roles and Future Visionary Endeavors of Current Emperor: Advancing Dynamic Methods for Longitudinal Microbiome Meta-Omics Data in Personalized and Precision Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400458. [PMID: 39535493 DOI: 10.1002/advs.202400458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 09/16/2024] [Indexed: 11/16/2024]
Abstract
Understanding the etiological complexity of diseases requires identifying biomarkers longitudinally associated with specific phenotypes. Advanced sequencing tools generate dynamic microbiome data, providing insights into microbial community functions and their impact on health. This review aims to explore the current roles and future visionary endeavors of dynamic methods for integrating longitudinal microbiome multi-omics data in personalized and precision medicine. This work seeks to synthesize existing research, propose best practices, and highlight innovative techniques. The development and application of advanced dynamic methods, including the unified analytical frameworks and deep learning tools in artificial intelligence, are critically examined. Aggregating data on microbes, metabolites, genes, and other entities offers profound insights into the interactions among microorganisms, host physiology, and external stimuli. Despite progress, the absence of gold standards for validating analytical protocols and data resources of various longitudinal multi-omics studies remains a significant challenge. The interdependence of workflow steps critically affects overall outcomes. This work provides a comprehensive roadmap for best practices, addressing current challenges with advanced dynamic methods. The review underscores the biological effects of clinical, experimental, and analytical protocol settings on outcomes. Establishing consensus on dynamic microbiome inter-studies and advancing reliable analytical protocols are pivotal for the future of personalized and precision medicine.
Collapse
Affiliation(s)
- Vera-Khlara S Oh
- Big Biomedical Data Integration and Statistical Analysis (DIANA) Research Center, Department of Data Science, College of Natural Sciences, Jeju National University, Jeju City, Jeju Do, 63243, South Korea
| | - Robert W Li
- United States Department of Agriculture, Agricultural Research Service, Animal Genomics and Improvement Laboratory, Beltsville, MD, 20705, USA
| |
Collapse
|
49
|
Tan R, Merrill C, Riley CF, Hammer MA, Kenney RT, Riley AA, Li J, Zink AC, Karl ST, Price KM, Sharabidze LK, Rowland SN, Bailey SJ, Stiemsma LT, Pennell A. Acute inorganic nitrate ingestion does not impact oral microbial composition, cognitive function, or high-intensity exercise performance in female team-sport athletes. Eur J Appl Physiol 2024; 124:3511-3525. [PMID: 39017740 PMCID: PMC11568988 DOI: 10.1007/s00421-024-05552-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
The purpose of this study was to investigate the effects of acute nitrate (NO3-)-rich beetroot juice ingestion on explosive and high-intensity exercise performance, oral microbiota composition, and cognitive flexibility (i.e., function), before and after maximal intermittent running exercise. Fifteen women team-sport athletes were assigned in a randomized, double-blind, crossover design to consume concentrated NO3--depleted beetroot juice (PL; 0.1 mmol NO3-) and NO3--rich beetroot juice (BR; 12.0 mmol NO3-) 2.5 h prior to performing a battery of exercise performance tasks and cognitive testing before and after the Yo-Yo intermittent recovery level 1 (YYIR1) running test. Resting plasma [NO3-] and plasma nitrite ([NO2-]) were elevated following BR (P < 0.001). BR did not impact global composition or relative abundance of taxa in the oral microbiome (P > 0.05) or cognitive flexibility before or after exercise (P > 0.05). There was no significant difference in performance during 20-m (PRE, PL: 4.38 ± 0.27 vs. BR: 4.38 ± 0.32 s; POST, PL: 4.45 ± 0.29 vs. BR: 4.43 ± 0.35 s) and 10-m sprints (PRE, PL 2.78 ± 0.15 vs. BR 2.79 ± 0.18 s; POST, PL: 2.82 ± 0.16 vs. BR: 2.81 ± 0.19 s), isokinetic handgrip dynamometry, medicine ball throw, horizontal countermovement jump, or YYIR1 (PL: 355 ± 163 m vs. BR: 368 ± 184 m) between BR and PL (P > 0.05). These findings indicate that acute dietary NO3- may not influence the oral microbiome, explosive and high-intensity exercise performance, or cognitive function in women team-sport athletes.
Collapse
Affiliation(s)
- Rachel Tan
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA.
| | - Courtney Merrill
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Chandler F Riley
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Maya A Hammer
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Ryan T Kenney
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Alyssa A Riley
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Jeffrey Li
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Alexandra C Zink
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Sean T Karl
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Katherine M Price
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Luka K Sharabidze
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Samantha N Rowland
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Stephen J Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Leah T Stiemsma
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| | - Adam Pennell
- Natural Science Division, Pepperdine University, Malibu, CA, 90263, USA
| |
Collapse
|
50
|
Amaral WZ, Kokroko N, Treangen TJ, Villapol S, Gomez-Pinilla F. Probiotic therapy modulates the brain-gut-liver microbiota axis in a mouse model of traumatic brain injury. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167483. [PMID: 39209236 PMCID: PMC11526848 DOI: 10.1016/j.bbadis.2024.167483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
The interplay between gut microbiota and host health is crucial for maintaining the overall health of the body and brain, and it is even more crucial how changes in the bacterial profile can influence the aftermath of traumatic brain injury (TBI). We studied the effects of probiotic treatment after TBI to identify potential changes in hepatic lipid species relevant to brain function. Bioinformatic analysis of the gut microbiota indicated a significant increase in the Firmicutes/Bacteroidetes ratio in the probiotic-treated TBI group compared to sham and untreated TBI groups. Although strong correlations between gut bacteria and hepatic lipids were found in sham mice, TBI disrupted these links, and probiotic treatment did not fully restore them. Probiotic treatment influenced systemic glucose metabolism, suggesting altered metabolic regulation. Behavioral tests confirmed memory improvement in probiotic-treated TBI mice. While TBI reduced hippocampal mRNA expression of CaMKII and CREB, probiotics reversed these effects yet did not alter BDNF mRNA levels. Elevated pro-inflammatory markers TNF-α and IL1-β in TBI mice were not significantly affected by probiotic treatment, pointing to different mechanisms underlying the probiotic benefits. In summary, our study suggests that TBI induces dysbiosis, alters hepatic lipid profiles, and preemptive administration of Lactobacillus helveticus and Bifidobacterium longum probiotics can counter neuroplasticity deficits and memory impairment. Altogether, these findings highlight the potential of probiotics for attenuating TBI's detrimental cognitive and metabolic effects through gut microbiome modulation and hepatic lipidomic alteration, laying the groundwork for probiotics as a potential TBI therapy.
Collapse
Affiliation(s)
- Wellington Z Amaral
- Departments of Neurosurgery and Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Natalie Kokroko
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Todd J Treangen
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Sonia Villapol
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Fernando Gomez-Pinilla
- Departments of Neurosurgery and Integrative Biology and Physiology, University of California, Los Angeles, CA, USA.
| |
Collapse
|