1
|
Iqbal M, Yu Q, Tang J, Xiang J. Unraveling the gut microbiota's role in obesity: key metabolites, microbial species, and therapeutic insights. J Bacteriol 2025; 207:e0047924. [PMID: 40183584 PMCID: PMC12096833 DOI: 10.1128/jb.00479-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Obesity, characterized by excessive fat accumulation, stems from an imbalance between energy intake and expenditure, with the gut microbiota playing a crucial role. This review highlights how gut microbiota influences metabolic pathways, inflammation, and adipose tissue regulation in obesity. Specific bacteria and metabolites, such as lipopolysaccharides (LPS) and short-chain fatty acids (SCFAs), modulate gut permeability, inflammation, and energy harvest, impacting obesity development. Certain gut bacteria, including Clostridium XIVb, Dorea spp., Enterobacter cloacae, and Collinsella aerofaciens, promote obesity by increasing energy harvest, gut permeability, and inflammatory response through LPS translocation into the bloodstream. Conversely, beneficial bacteria like Akkermansia muciniphila, Lactobacillus spp., and Bifidobacterium spp. enhance gut barrier integrity, regulate SCFA production, and modulate fasting-induced adipose factor, which collectively support metabolic health by reducing fat storage and inflammation. Metabolites such as SCFAs (acetate, propionate, and butyrate) interact with G-protein coupled receptors to regulate lipid metabolism and promote the browning of white adipose tissue (WAT), thus enhancing thermogenesis and energy expenditure. However, LPS contributes to insulin resistance and fat accumulation, highlighting the dual roles of these microbial metabolites in both supporting and disrupting metabolic function. Therapeutic interventions targeting gut microbiota, such as promoting WAT browning and activating brown adipose tissue (BAT), hold promise for obesity management. However, personalized approaches are necessary due to individual microbiome variability. Further research is essential to translate these insights into microbiota-based clinical therapies.
Collapse
Affiliation(s)
- Majid Iqbal
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Yu
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingqun Tang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juanjuan Xiang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Zhao J, Miao D. Precision oncology in colorectal cancer: An anatomical revolution through molecular-clinical integration across colonic subsites. Clin Res Hepatol Gastroenterol 2025; 49:102613. [PMID: 40374162 DOI: 10.1016/j.clinre.2025.102613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/08/2025] [Accepted: 05/10/2025] [Indexed: 05/17/2025]
Abstract
Colorectal cancer (CRC) exhibits significant heterogeneity across different colonic subsites, which vary in embryological origin, microbiome, metabolome, and molecular profiles, affecting tumorigenesis, treatment response, and prognosis. We emphasize the importance of this subsite heterogeneity to advance precision medicine in CRC.
Collapse
Affiliation(s)
- Jiefeng Zhao
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China.
| | - Daxing Miao
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China.
| |
Collapse
|
3
|
Lyte JM, Seyoum MM, Ayala D, Kers JG, Caputi V, Johnson T, Zhang L, Rehberger J, Zhang G, Dridi S, Hale B, De Oliveira JE, Grum D, Smith AH, Kogut M, Ricke SC, Ballou A, Potter B, Proszkowiec-Weglarz M. Do we need a standardized 16S rRNA gene amplicon sequencing analysis protocol for poultry microbiota research? Poult Sci 2025; 104:105242. [PMID: 40334389 DOI: 10.1016/j.psj.2025.105242] [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: 01/24/2025] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/09/2025] Open
Abstract
Bacteria are the major component of poultry gastrointestinal tract (GIT) microbiota and play an important role in host health, nutrition, physiology regulation, intestinal development, and growth. Bacterial community profiling based on the 16S ribosomal RNA (rRNA) gene amplicon sequencing approach has become the most popular method to determine the taxonomic composition and diversity of the poultry microbiota. The 16S rRNA gene profiling involves numerous steps, including sample collection and storage, DNA isolation, 16S rRNA gene primer selection, Polymerase Chain Reaction (PCR), library preparation, sequencing, raw sequencing reads processing, taxonomic classification, α- and β-diversity calculations, and statistical analysis. However, there is currently no standardized protocol for 16S rRNA gene analysis profiling and data deposition for poultry microbiota studies. Variations in DNA storage and isolation, primer design, and library preparation are known to introduce biases, affecting community structure and microbial population analysis leading to over- or under-representation of individual bacteria within communities. Additionally, different sequencing platforms, bioinformatics pipeline, and taxonomic database selection can affect classification and determination of the microbial taxa. Moreover, detailed experimental design and DNA processing and sequencing methods are often inadequately reported in poultry 16S rRNA gene sequencing studies. Consequently, poultry microbiota results are often difficult to reproduce and compare across studies. This manuscript reviews current practices in profiling poultry microbiota using 16S rRNA gene amplicon sequencing and proposes the development of guidelines for protocol for 16S rRNA gene sequencing that spans from sample collection through data deposition to achieve more reliable data comparisons across studies and allow for comparisons and/or interpretations of poultry studies conducted worldwide.
Collapse
Affiliation(s)
- Joshua M Lyte
- United States Department of Agriculture, Agricultural Research Service, Southeast Area, Poultry Production and Product Safety Research, Fayetteville 72701, AR, United States
| | - Mitiku M Seyoum
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville 72701, AR, United States
| | - Diana Ayala
- Purina Animal Nutrition Center, Land O'Lakes, Gray Summit 63039, MO, United States
| | - Jannigje G Kers
- Faculty of Veterinary Medicine, Utrecht University, and Laboratory of Microbiology, Wageningen University & Research, The Netherlands
| | - Valentina Caputi
- United States Department of Agriculture, Agricultural Research Service, Southeast Area, Poultry Production and Product Safety Research, Fayetteville 72701, AR, United States
| | - Timothy Johnson
- University of Minnesota, Saint Paul 55108, MN, United States
| | - Li Zhang
- Mississippi State University, Mississippi State 39762, MS, United States
| | - Joshua Rehberger
- Arm and Hammer Animal Nutrition, Waukesha 53186, WI, United States
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater 74078, OK, United States
| | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville 72701, AR, United States
| | - Brett Hale
- AgriGro, Doniphan 6393, MO, United States
| | | | - Daniel Grum
- Purina Animal Nutrition Center, Land O'Lakes, Gray Summit 63039, MO, United States
| | - Alexandra H Smith
- Mississippi State University, Mississippi State 39762, MS, United States
| | - Michael Kogut
- United States Department of Agriculture, Agricultural Research Service, Southern Plains Agricultural Research Center, College Station 77845, TX, United States
| | - Steven C Ricke
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, 53706, WI, United States
| | - Anne Ballou
- Iluma Alliance, Durham 27703, NC, United States
| | - Bill Potter
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville 72701, AR, United States
| | - Monika Proszkowiec-Weglarz
- United States Department of Agriculture, Agricultural Research Service, Northeast Area, Beltsville Agriculture Research Center, Animal Biosciences and Biotechnology Laboratory, Beltsville 20705, MD, United States.
| |
Collapse
|
4
|
Tronel A, Roger-Margueritat M, Plazy C, Biennier S, Craspay A, Mohanty I, Portier SC, Laiola M, Roeselers G, Mathieu N, Hupe M, Dorrestein PC, Alcaraz JP, Martin D, Cinquin P, Silvent AS, Giai J, Proust M, Soranzo T, Buelow E, Gouellec ALE. Profiling the human luminal small intestinal microbiome using a novel ingestible medical device. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.18.25326056. [PMID: 40321269 PMCID: PMC12047917 DOI: 10.1101/2025.04.18.25326056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
The invasive nature of sample collection for studying the small intestinal (SI) microbiome often results in its poor characterization. This study evaluated a novel ingestible medical device (MD) for SI luminal sample collection. A monocentric interventional trial (NCT05477069) was conducted on 15 healthy subjects. Metagenomics, metabolomics and culturomics assessed the MD's effectiveness in characterizing the healthy SI microbiome and identifying potential biomarkers. The SI microbiota differed significantly from the fecal microbiota, displaying high inter-individual variability, lower species richness, and reduced alpha diversity. A combined untargeted and semi-targeted LC-MS/MS metabolomics approach identified a distinct SI metabolic footprint, with bile acids and amino acids being the most abundant classes of metabolites. Host and host/microbe-derived bile acids were particularly abundant in SI samples. The application of a fast culturomics approach to two SI samples enabled species-level characterization, resulting in the identification of 90 bacterial species, including five potential novel species. The present study demonstrates the efficacy of our novel sampling MD in enabling comprehensive SI microbiome analysis through an integrative multi-omics approach, allowing the identification of distinct microbiome signatures between SI and fecal samples.
Collapse
Affiliation(s)
- Alexandre Tronel
- Pelican Health, 5 avenue du Grand Sablon, 38700, La Tronche, France
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Morgane Roger-Margueritat
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Caroline Plazy
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
- Service de Biochimie Biologie Moléculaire Toxicologie Environnementale, UM Biochimie des Enzymes et des Protéines, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
- Plateforme de Métabolomique GEMELI-GExiM, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
| | - Salomé Biennier
- Pelican Health, 5 avenue du Grand Sablon, 38700, La Tronche, France
| | - Anthony Craspay
- Pelican Health, 5 avenue du Grand Sablon, 38700, La Tronche, France
| | - Ipsita Mohanty
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Stéphanie Cools Portier
- Danone Global Research & Innovation Center, Route départementale 128, 91 190 Gif sur Yvette, France
| | - Manolo Laiola
- Danone Global Research & Innovation Center, Route départementale 128, 91 190 Gif sur Yvette, France
| | - Guus Roeselers
- Danone Global Research & Innovation Center, Route départementale 128, 91 190 Gif sur Yvette, France
| | - Nicolas Mathieu
- Univ. Grenoble Alpes/Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes/Institute for Advanced Biosciences, CNRS UMR 5309-INSERM U1209, 38043 Grenoble, France
| | - Marianne Hupe
- Univ. Grenoble Alpes/Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes/Institute for Advanced Biosciences, CNRS UMR 5309-INSERM U1209, 38043 Grenoble, France
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jean-Pierre Alcaraz
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Donald Martin
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Philippe Cinquin
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Anne-Sophie Silvent
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CIC, 38000 Grenoble, France
| | - Joris Giai
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CIC, 38000 Grenoble, France
| | - Marion Proust
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CIC, 38000 Grenoble, France
| | - Thomas Soranzo
- Pelican Health, 5 avenue du Grand Sablon, 38700, La Tronche, France
| | - Elena Buelow
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Audrey LE Gouellec
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
- Service de Biochimie Biologie Moléculaire Toxicologie Environnementale, UM Biochimie des Enzymes et des Protéines, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
- Plateforme de Métabolomique GEMELI-GExiM, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
| |
Collapse
|
5
|
Yang K, Li G, Li Q, Wang W, Zhao X, Shao N, Qiu H, Liu J, Xu L, Zhao J. Distribution of gut microbiota across intestinal segments and their impact on human physiological and pathological processes. Cell Biosci 2025; 15:47. [PMID: 40241220 PMCID: PMC12001467 DOI: 10.1186/s13578-025-01385-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
In recent years, advancements in metagenomics, metabolomics, and single-cell sequencing have enhanced our understanding of the intricate relationships between gut microbiota and their hosts. Gut microbiota colonize humans from birth, with their initial composition significantly influenced by the mode of delivery and feeding method. During the transition from infancy to early childhood, exposure to a diverse diet and the maturation of the immune system lead to the gradual stabilization of gut microbiota's composition and distribution. Numerous studies have demonstrated that gut microbiota can influence a wide range of physiological functions and pathological processes by interacting with various tissues and organs through the gut-organ axis. Different intestinal segments exhibit unique physical and chemical conditions, which leads to the formation of vertical gradients along the intestinal tract: aerobes and facultative aerobes mainly live in the small intestine and anaerobic bacteria mainly live in the large intestine, and horizontal gradients: mucosa-associated microbiota and lumen-associated microbiota. In this review, we systematically summarize the distribution characteristics of gut microbiota across six intestinal segments: duodenum, jejunum, ileum, cecum, colon, and rectum. We also draw a conclusion that gut microbiota distributed in different intestinal segments affect the progression of different diseases. We hope to elucidate the role of microbiota at specific anatomic sites within the gut in precisely regulating the processes of particular diseases, thereby providing a solid foundation for developing novel diagnostic and therapeutic strategies for related diseases.
Collapse
Affiliation(s)
- Ke Yang
- The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Guangqin Li
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Qihong Li
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Wei Wang
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xu Zhao
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Guizhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Nan Shao
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Hui Qiu
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Jing Liu
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Lin Xu
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| | - Juanjuan Zhao
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
6
|
Parsons BW, Bedford MR, Wyatt CL. Evaluation of the incremental effect of fasting length on pH, select cecal microbial groups, cecal volatile fatty acid concentrations, and secretory IgA excretion in roosters. Poult Sci 2025; 104:105161. [PMID: 40273682 PMCID: PMC12051570 DOI: 10.1016/j.psj.2025.105161] [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: 01/23/2025] [Revised: 04/05/2025] [Accepted: 04/13/2025] [Indexed: 04/26/2025] Open
Abstract
Two experiments were conducted to evaluate the effect of incremental fasting time on the gastrointestinal tract in chickens. Adult White Leghorn roosters with intact ceca were provided a nutrient-adequate corn-soybean meal diet ad libitum for 3 weeks. Prior to initiation of the experimental phase, ad libitum feed intake was recorded for 8 h and immediately after the fasting period commenced. In Experiment 1, roosters were fasted for either 0, 3, 6, 9, 12, 16, or 24 h. At each time point birds were euthanized and pH in the crop, gizzard, and ceca were recorded and cecal contents were collected to measure volatile fatty acids (VFA) and select cecal microbial groups. In Experiment 2, roosters were fasted for 0, 3, 6, 9, 12, 16, and 24 h and excreta were collected to determine secretory IgA (sIgA) excretion. In contrast to Experiment 1, roosters in Experiment 2 were not euthanized and thus sIgA excretion was measured within individual roosters across each time point. Experiments 1 and 2 contained 5 and 8 replicates per treatment, respectively. In Experiment 1, there was a linear increase (P < 0.05) in cecal pH as fasting length increased. Cecal VFA content was reduced (P < 0.05) by 9 to 12 h of fasting and branch-chain FA to VFA ratio increased (P < 0.05) by 6 h of fasting. There were few effects (P > 0.05) of fasting on the microbial groups in cecal contents and mucosa; however, Escherichia coli content was greater (P < 0.05) at 24 h of fasting compared with other time points. In Experiment 2, total sIgA excreted was greater (P < 0.05) at 24 h of fasting, being 1106 µg/h at 24 h compared with a mean of 419 µg/h for all other time points. In conclusion, fasting reduced cecal VFA concentrations and increased cecal pH, Escherichia coli, branched-chain FA to VFA ratio, and sIgA excretion, suggesting that fasting elicited negative effects on the gastrointestinal tract.
Collapse
Affiliation(s)
- B W Parsons
- Department of Poultry Science, University of Arkansas at Fayetteville, AR 72701, USA.
| | | | - C L Wyatt
- AB Vista, Marlborough, United Kingdom
| |
Collapse
|
7
|
Hanauska A, Sommerfeld V, Schollenberger M, Huber K, Rodehutscord M. Endogenous mucosal phosphatases characterization in duodenum brush border membrane of laying hens. Front Physiol 2025; 16:1581088. [PMID: 40241716 PMCID: PMC11999836 DOI: 10.3389/fphys.2025.1581088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Chicken mucosal phosphatases can partially degrade phytate contained in the feed. Little is known about the characteristics and degradation products of such mucosal phosphatases and the effects of age and genetic strain of the chicken. The objective of this study was to characterize endogenous mucosal phosphatases of two laying hen strains fed diets with or without mineral phosphorus (P) before and after the onset of egg laying. Hens of the strains Lohmann Brown-classic (LB) and Lohmann LSL-classic (LSL) were sacrificed in weeks 19 and 24 of age after 4 weeks of feeding one of two diets with (P+) or without (P-) mineral P supplement. Mucosa of the duodenum was collected, and the brush border membrane (BBM) of enterocytes was enriched and used for phosphatase activity determination. Additionally, the BBM was used in a modified three-step in vitro assay to study the InsP6 degradation products. The results of both in vitro assays were not significantly affected by hen strain and diet. The activity of mucosal phosphatase in 19-week-old hens was, on average, 0.8 µmol Pi/g BBM protein/min lower than in 24-week-old hens (P < 0.002). Consistently, the InsP6 concentration in the incubation residue was significantly higher in 19-week-old hens than in 24-week-old hens (P < 0.001). In the incubation residue, the concentrations of Ins(1,2,3,4,5)P5, Ins(1,2,3,4,6)P5, and Ins(1,2,3,4)P4 were significantly lower (P ≤ 0.002), and those of InsP3 and InsP2 were significantly higher (P ≤ 0.027) when BBM of 24-week-old hens was used compared to 19-week-old hens. The InsP6 degradation products suggest the activity primarily of a 6- and secondarily of a 5-phytase in the duodenal mucosa. The consistent results from both in vitro assays provide a comprehensive characterization of these enzymes. Under the conditions of this study, small intestine calcium concentration appeared to influence mucosal enzyme activity more than dietary mineral P supplementation.
Collapse
|
8
|
Fruet C, Müller EL, Loverdo C, Bitbol AF. Spatial structure facilitates evolutionary rescue by drug resistance. PLoS Comput Biol 2025; 21:e1012861. [PMID: 40179127 PMCID: PMC11967957 DOI: 10.1371/journal.pcbi.1012861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/09/2025] [Indexed: 04/05/2025] Open
Abstract
Bacterial populations often have complex spatial structures, which can impact their evolution. Here, we study how spatial structure affects the evolution of antibiotic resistance in a bacterial population. We consider a minimal model of spatially structured populations where all demes (i.e., subpopulations) are identical and connected to each other by identical migration rates. We show that spatial structure can facilitate the survival of a bacterial population to antibiotic treatment, starting from a sensitive inoculum. Specifically, the bacterial population can be rescued if antibiotic resistant mutants appear and are present when drug is added, and spatial structure can impact the fate of these mutants and the probability that they are present. Indeed, the probability of fixation of neutral or deleterious mutations providing drug resistance is increased in smaller populations. This promotes local fixation of resistant mutants in the structured population, which facilitates evolutionary rescue by drug resistance in the rare mutation regime. Once the population is rescued by resistance, migrations allow resistant mutants to spread in all demes. Our main result that spatial structure facilitates evolutionary rescue by antibiotic resistance extends to more complex spatial structures, and to the case where there are resistant mutants in the inoculum.
Collapse
Affiliation(s)
- Cecilia Fruet
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| | - Ella Linxia Müller
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| | - Claude Loverdo
- Sorbonne Université, CNRS,Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), Paris,France
| | - Anne-Florence Bitbol
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
9
|
Soh M, Er S, Low A, Jaafar Z, de Boucher R, Seedorf H. Spatial and temporal changes in gut microbiota composition of farmed Asian seabass ( Lates calcarifer) in different aquaculture settings. Microbiol Spectr 2025; 13:e0198924. [PMID: 40084873 PMCID: PMC12054105 DOI: 10.1128/spectrum.01989-24] [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: 08/07/2024] [Accepted: 02/09/2025] [Indexed: 03/16/2025] Open
Abstract
The microbiota composition of healthy farmed fishes remains poorly characterized for many species. This study explores the influence of the external environment and innate factors that may shape the gut microbiota of farmed Asian seabass, Lates calcarifer. The α-diversity based on Shannon, Simpson, and Chao1 indices was lower for fishes reared in sea cages and tanks than for fishes that experienced a transfer from sea cages to tanks. Longitudinal analyses of gut segments revealed no significant differences in alpha diversity between segments within the same containment type, except for the Chao1 index between the stomach and pyloric cecum of sea-caged fishes. β-diversity analysis using weighted UniFrac distance and Bray-Curtis dissimilarity demonstrated that fish reared in the same containment type shared similar microbial communities. PERMANOVA tests confirmed that containment type, farm, and batch significantly influenced these distances. Containment type accounted for 10.4% of the observed diversity, farm for 29.8%, and batch for 10.7%. Genera comprising potential pathogens such as Aeromonas, Flavobacterium, and Vibrio were differentially abundant along the guts of fish from different containment types and particularly increased in tanks. Microbiota changes were observed with host age and gut segment, with differentially abundant microbial genera identified along the gut and as the seabass grew. Comparing the hindgut microbiota of Asian seabass to other species of farmed fishes revealed host-specific clustering as indicated by PERMANOVA. Overall, these findings underscore the significance of containment conditions on the gut microbiota of Asian seabass, with broad implications for aquaculture practices. IMPORTANCE Understanding the microbiota composition of healthy farmed fishes is crucial for optimizing aquaculture practices. This study highlights the significant influence of containment conditions on the gut microbiota of farmed Asian seabass (Lates calcarifer). By demonstrating that gut microbiota diversity and community composition are shaped by containment type, farm location, and batch, the research provides valuable insights into how external environmental factors and innate host factors interact to influence fish health. The findings, particularly the differential abundance of potential pathogens in various containment types, underscore the need for tailored management strategies in aquaculture. This research not only advances our knowledge of fish microbiota but also has broad implications for improving the sustainability and productivity of aquaculture practices.
Collapse
Affiliation(s)
- Melissa Soh
- Temasek Life Sciences Laboratory, Singapore, Singapore
| | - Shuan Er
- Temasek Life Sciences Laboratory, Singapore, Singapore
| | - Adrian Low
- Temasek Life Sciences Laboratory, Singapore, Singapore
| | - Zeehan Jaafar
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | | | - Henning Seedorf
- Temasek Life Sciences Laboratory, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
10
|
Yang JC, Lagishetty V, Aja E, Arias-Jayo N, Chang C, Hauer M, Katzka W, Zhou Y, Sedighian F, Koletic C, Liang F, Dong TS, Situ J, Troutman R, Buri H, Bhute S, Simpson CA, Braun J, Jacob N, Jacobs JP. Biogeographical distribution of gut microbiome composition and function is partially recapitulated by fecal transplantation into germ-free mice. THE ISME JOURNAL 2025; 19:wrae250. [PMID: 39680691 PMCID: PMC11973428 DOI: 10.1093/ismejo/wrae250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/24/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Fecal microbiota transplantation has been vital for establishing whether host phenotypes can be conferred through the microbiome. However, whether the existing microbial ecology along the mouse gastrointestinal tract can be recapitulated in germ-free mice colonized with stool remains unknown. We first identified microbes and their predicted functions specific to each of six intestinal regions in three cohorts of specific pathogen-free mice spanning two facilities. Of these region-specific microbes, the health-linked genus Akkermansia was consistently enriched in the lumen of the small intestine compared to the colon. Predictive functional modeling on 16S rRNA gene amplicon sequencing data recapitulated in shotgun sequencing data revealed increased microbial central metabolism, lipolytic fermentation, and cross-feeding in the small intestine, whereas butyrate synthesis was colon-enriched. Neuroactive compound metabolism also demonstrated regional specificity, including small intestine-enriched gamma-aminobutyric acid degradation and colon-enriched tryptophan degradation. Specifically, the jejunum and ileum stood out as sites with high predicted metabolic and neuromodulation activity. Differences between luminal and mucosal microbiomes within each site of the gastrointestinal tract were largely facility-specific, though there were a few consistent patterns in microbial metabolism in specific pathogen-free mice. These included luminal enrichment of central metabolism and cross-feeding within both the small intestine and the colon, and mucosal enrichment of butyrate synthesis within the colon. Across three cohorts of germ-free mice colonized with mice or human stool, compositional and functional region specificity were inconsistently reproduced. These results underscore the importance of investigating the spatial variation of the gut microbiome to better understand its impact on host physiology.
Collapse
Affiliation(s)
- Julianne C Yang
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Venu Lagishetty
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Ezinne Aja
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
- Goodman-Luskin Microbiome Center at UCLA, Center for Health Sciences 42-210, 650 Charles E. Young Dr. S., Los Angeles, CA 90095-7378, United States
| | - Nerea Arias-Jayo
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Candace Chang
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Megan Hauer
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - William Katzka
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
- Goodman-Luskin Microbiome Center at UCLA, Center for Health Sciences 42-210, 650 Charles E. Young Dr. S., Los Angeles, CA 90095-7378, United States
| | - Yi Zhou
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
- West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, PR China
| | - Farzaneh Sedighian
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, 11301 Wilshire Boulevard, Los Angeles, CA 90073-1003, United States
| | - Carolina Koletic
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Fengting Liang
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Tien S Dong
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, 11301 Wilshire Boulevard, Los Angeles, CA 90073-1003, United States
| | - Jamilla Situ
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Ryan Troutman
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Heidi Buri
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Shrikant Bhute
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
- Goodman-Luskin Microbiome Center at UCLA, Center for Health Sciences 42-210, 650 Charles E. Young Dr. S., Los Angeles, CA 90095-7378, United States
| | - Carra A Simpson
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
| | - Jonathan Braun
- F. Widjaja Foundation Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048-1865, United States
| | - Noam Jacob
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, 11301 Wilshire Boulevard, Los Angeles, CA 90073-1003, United States
- F. Widjaja Foundation Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048-1865, United States
| | - Jonathan P Jacobs
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, 200 Medical Plaza, Los Angeles, CA 90024-2484, United States
- Goodman-Luskin Microbiome Center at UCLA, Center for Health Sciences 42-210, 650 Charles E. Young Dr. S., Los Angeles, CA 90095-7378, United States
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, 11301 Wilshire Boulevard, Los Angeles, CA 90073-1003, United States
| |
Collapse
|
11
|
Hoffmann SV, O'Shea JP, Galvin P, Jannin V, Griffin BT. State-of-the-art and future perspectives in ingestible remotely controlled smart capsules for drug delivery: A GENEGUT review. Eur J Pharm Sci 2024; 203:106911. [PMID: 39293502 DOI: 10.1016/j.ejps.2024.106911] [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: 07/05/2024] [Revised: 09/06/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
An emerging concern globally, particularly in developed countries, is the rising prevalence of Inflammatory Bowel Disease (IBD), such as Crohn's disease. Oral delivery technologies that can release the active therapeutic cargo specifically at selected sites of inflammation offer great promise to maximise treatment outcomes and minimise off-target effects. Therapeutic strategies for IBD have expanded in recent years, with an increasing focus on biologic and nucleic acid-based therapies. Reliable site-specific delivery in the gastrointestinal (GI) tract is particularly crucial for these therapeutics to ensure sufficient concentrations in the targeted cells. Ingestible smart capsules hold great potential for precise drug delivery. Despite previous unsuccessful endeavours to commercialise drug delivery smart capsules, the current rise in demand and recent advancements in component development, manufacturing, and miniaturisation have reignited interest in ingestible devices. Consequently, this review analyses the advancements in various mechanical and electrical components associated with ingestible smart drug delivery capsules. These components include modules for device localisation, actuation and retention within the GI tract, signal transmission, drug release, power supply, and payload storage. Challenges and constraints associated with previous capsule design functionality are presented, followed by a critical outlook on future design considerations to ensure efficient and reliable site-specific delivery for the local treatment of GI disorders.
Collapse
Affiliation(s)
- Sophia V Hoffmann
- School of Pharmacy, University College Cork, College Road, Cork, Ireland
| | - Joseph P O'Shea
- School of Pharmacy, University College Cork, College Road, Cork, Ireland
| | - Paul Galvin
- Tyndall National Institute, University College Cork, Cork T12R5CP, Ireland
| | | | - Brendan T Griffin
- School of Pharmacy, University College Cork, College Road, Cork, Ireland.
| |
Collapse
|
12
|
Nagpal S, Srivastava SK. Colon or semicolon: gut sampling microdevices for omics insights. NPJ Biofilms Microbiomes 2024; 10:97. [PMID: 39358351 PMCID: PMC11447266 DOI: 10.1038/s41522-024-00536-2] [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: 12/20/2023] [Accepted: 07/19/2024] [Indexed: 10/04/2024] Open
Abstract
Ingestible microdevices represent a breakthrough in non-invasive sampling of the human gastrointestinal (GI) tract. By capturing the native spatiotemporal microbiome and intricate biochemical gradients, these devices allow a non-invasive multi-omic access to the unperturbed host-microbiota crosstalk, immune/nutritional landscapes and gut-organ connections. We present the current progress of GI sampling microdevices towards personalized metabolism and fostering collaboration among clinicians, engineers, and data scientists.
Collapse
Affiliation(s)
- Sunil Nagpal
- TCS Research, Tata Consultancy Services Ltd, Pune, India
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Sarvesh Kumar Srivastava
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India.
- Department of Biomedical Engineering, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
13
|
Dallavilla T, Galiè S, Sambruni G, Borin S, Fazio N, Fumagalli-Romario U, Manzo T, Nezi L, Schaefer MH. Differences in the molecular organisation of tumours along the colon are linked to interactions within the tumour ecosystem. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167311. [PMID: 38909851 DOI: 10.1016/j.bbadis.2024.167311] [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: 03/08/2024] [Revised: 05/22/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
Tumours exhibit significant heterogeneity in their molecular profiles across patients, largely influenced by the tissue of origin, where certain driver gene mutations are predominantly associated with specific cancer types. Here, we unveil an additional layer of complexity: some cancer types display anatomic location-specific mutation profiles akin to tissue-specificity. To better understand this phenomenon, we concentrate on colon cancer. While prior studies have noted changes of the frequency of molecular alterations along the colon, the underlying reasons and whether those changes occur rather gradual or are distinct between the left and right colon, remain unclear. Developing and leveraging stringent statistical models on molecular data from 522 colorectal tumours from The Cancer Genome Atlas, we reveal disparities in molecular properties between the left and right colon affecting many genes. Interestingly, alterations in genes responsive to environmental cues and properties of the tumour ecosystem, including metabolites which we quantify in a cohort of 27 colorectal cancer patients, exhibit continuous trends along the colon. Employing network methodologies, we uncover close interactions between metabolites and genes, including drivers of colon cancer, showing continuous abundance or alteration profiles. This underscores how anatomic biases in the composition and interactions within the tumour ecosystem help explaining gradients of carcinogenesis along the colon.
Collapse
Affiliation(s)
- Tiziano Dallavilla
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Milano, Italy
| | - Serena Galiè
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Milano, Italy
| | - Gaia Sambruni
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Milano, Italy
| | - Simona Borin
- Digestive Surgery, European Institute of Oncology-IRCCS, Milano, Italy
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology-IRCCS, Milano, Italy
| | | | - Teresa Manzo
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Milano, Italy
| | - Luigi Nezi
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Milano, Italy
| | - Martin H Schaefer
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Milano, Italy.
| |
Collapse
|
14
|
Timmermans S, Wallaeys C, Garcia-Gonzalez N, Pollaris L, Saeys Y, Libert C. Identification and Characterization of Multiple Paneth Cell Types in the Mouse Small Intestine. Cells 2024; 13:1435. [PMID: 39273007 PMCID: PMC11394207 DOI: 10.3390/cells13171435] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The small intestinal crypts harbor secretory Paneth cells (PCs) which express bactericidal peptides that are crucial for maintaining intestinal homeostasis. Considering the diverse environmental conditions throughout the course of the small intestine, multiple subtypes of PCs are expected to exist. We applied single-cell RNA-sequencing of PCs combined with deep bulk RNA-sequencing on PC populations of different small intestinal locations and discovered several expression-based PC clusters. Some of these are discrete and resemble tuft cell-like PCs, goblet cell (GC)-like PCs, PCs expressing stem cell markers, and atypical PCs. Other clusters are less discrete but appear to be derived from different locations along the intestinal tract and have environment-dictated functions such as food digestion and antimicrobial peptide production. A comprehensive spatial analysis using Resolve Bioscience was conducted, leading to the identification of different PC's transcriptomic identities along the different compartments of the intestine, but not between PCs in the crypts themselves.
Collapse
Affiliation(s)
- Steven Timmermans
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; (S.T.); (C.W.); (N.G.-G.); (L.P.); (Y.S.)
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Charlotte Wallaeys
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; (S.T.); (C.W.); (N.G.-G.); (L.P.); (Y.S.)
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Natalia Garcia-Gonzalez
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; (S.T.); (C.W.); (N.G.-G.); (L.P.); (Y.S.)
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Lotte Pollaris
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; (S.T.); (C.W.); (N.G.-G.); (L.P.); (Y.S.)
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000 Ghent, Belgium
| | - Yvan Saeys
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; (S.T.); (C.W.); (N.G.-G.); (L.P.); (Y.S.)
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000 Ghent, Belgium
| | - Claude Libert
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; (S.T.); (C.W.); (N.G.-G.); (L.P.); (Y.S.)
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| |
Collapse
|
15
|
Sastre DE, Sultana N, V A S Navarro M, Huliciak M, Du J, Cifuente JO, Flowers M, Liu X, Lollar P, Trastoy B, Guerin ME, Sundberg EJ. Human gut microbes express functionally distinct endoglycosidases to metabolize the same N-glycan substrate. Nat Commun 2024; 15:5123. [PMID: 38879612 PMCID: PMC11180146 DOI: 10.1038/s41467-024-48802-3] [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: 12/12/2023] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
Bacteroidales (syn. Bacteroidetes) are prominent members of the human gastrointestinal ecosystem mainly due to their efficient glycan-degrading machinery, organized into gene clusters known as polysaccharide utilization loci (PULs). A single PUL was reported for catabolism of high-mannose (HM) N-glycan glyco-polypeptides in the gut symbiont Bacteroides thetaiotaomicron, encoding a surface endo-β-N-acetylglucosaminidase (ENGase), BT3987. Here, we discover an ENGase from the GH18 family in B. thetaiotaomicron, BT1285, encoded in a distinct PUL with its own repertoire of proteins for catabolism of the same HM N-glycan substrate as that of BT3987. We employ X-ray crystallography, electron microscopy, mass spectrometry-based activity measurements, alanine scanning mutagenesis and a broad range of biophysical methods to comprehensively define the molecular mechanism by which BT1285 recognizes and hydrolyzes HM N-glycans, revealing that the stabilities and activities of BT1285 and BT3987 were optimal in markedly different conditions. BT1285 exhibits significantly higher affinity and faster hydrolysis of poorly accessible HM N-glycans than does BT3987. We also find that two HM-processing endoglycosidases from the human gut-resident Alistipes finegoldii display condition-specific functional properties. Altogether, our data suggest that human gut microbes employ evolutionary strategies to express distinct ENGases in order to optimally metabolize the same N-glycan substrate in the gastroinstestinal tract.
Collapse
Affiliation(s)
- Diego E Sastre
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA.
| | - Nazneen Sultana
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Structural Biochemistry Unit, National Institute of Dental and Craniofacial Research (NIDCR/NIH), Bethesda, MD, USA
| | - Marcos V A S Navarro
- Institute of Physics (IFSC-USP), University of São Paulo, São Carlos, SP, Brazil
- Center for Innovative Proteomics, Cornell University, Ithaca, NY, USA
| | - Maros Huliciak
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Javier O Cifuente
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain
| | - Maria Flowers
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Xu Liu
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Pete Lollar
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Beatriz Trastoy
- Structural Glycoimmunology Laboratory, Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Marcelo E Guerin
- Structural Glycobiology Laboratory, Department of Structural and Molecular Biology, Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), Barcelona Science Park, c/Baldiri Reixac 4-8, Tower R, Barcelona, Catalonia, Spain
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
16
|
Freire TFA, Hu Z, Wood KB, Gjini E. Modeling spatial evolution of multi-drug resistance under drug environmental gradients. PLoS Comput Biol 2024; 20:e1012098. [PMID: 38820350 PMCID: PMC11142541 DOI: 10.1371/journal.pcbi.1012098] [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: 11/17/2023] [Accepted: 04/23/2024] [Indexed: 06/02/2024] Open
Abstract
Multi-drug combinations to treat bacterial populations are at the forefront of approaches for infection control and prevention of antibiotic resistance. Although the evolution of antibiotic resistance has been theoretically studied with mathematical population dynamics models, extensions to spatial dynamics remain rare in the literature, including in particular spatial evolution of multi-drug resistance. In this study, we propose a reaction-diffusion system that describes the multi-drug evolution of bacteria based on a drug-concentration rescaling approach. We show how the resistance to drugs in space, and the consequent adaptation of growth rate, is governed by a Price equation with diffusion, integrating features of drug interactions and collateral resistances or sensitivities to the drugs. We study spatial versions of the model where the distribution of drugs is homogeneous across space, and where the drugs vary environmentally in a piecewise-constant, linear and nonlinear manner. Although in many evolution models, per capita growth rate is a natural surrogate for fitness, in spatially-extended, potentially heterogeneous habitats, fitness is an emergent property that potentially reflects additional complexities, from boundary conditions to the specific spatial variation of growth rates. Applying concepts from perturbation theory and reaction-diffusion equations, we propose an analytical metric for characterization of average mutant fitness in the spatial system based on the principal eigenvalue of our linear problem, λ1. This enables an accurate translation from drug spatial gradients and mutant antibiotic susceptibility traits to the relative advantage of each mutant across the environment. Our approach allows one to predict the precise outcomes of selection among mutants over space, ultimately from comparing their λ1 values, which encode a critical interplay between growth functions, movement traits, habitat size and boundary conditions. Such mathematical understanding opens new avenues for multi-drug therapeutic optimization.
Collapse
Affiliation(s)
- Tomas Ferreira Amaro Freire
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Zhijian Hu
- Departments of Biophysics and Physics, University of Michigan, United States of America
| | - Kevin B. Wood
- Departments of Biophysics and Physics, University of Michigan, United States of America
| | - Erida Gjini
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
17
|
Cani PD, Van Hul M. Gut microbiota in overweight and obesity: crosstalk with adipose tissue. Nat Rev Gastroenterol Hepatol 2024; 21:164-183. [PMID: 38066102 DOI: 10.1038/s41575-023-00867-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 03/02/2024]
Abstract
Overweight and obesity are characterized by excessive fat mass accumulation produced when energy intake exceeds energy expenditure. One plausible way to control energy expenditure is to modulate thermogenic pathways in white adipose tissue (WAT) and/or brown adipose tissue (BAT). Among the different environmental factors capable of influencing host metabolism and energy balance, the gut microbiota is now considered a key player. Following pioneering studies showing that mice lacking gut microbes (that is, germ-free mice) or depleted of their gut microbiota (that is, using antibiotics) developed less adipose tissue, numerous studies have investigated the complex interactions existing between gut bacteria, some of their membrane components (that is, lipopolysaccharides), and their metabolites (that is, short-chain fatty acids, endocannabinoids, bile acids, aryl hydrocarbon receptor ligands and tryptophan derivatives) as well as their contribution to the browning and/or beiging of WAT and changes in BAT activity. In this Review, we discuss the general physiology of both WAT and BAT. Subsequently, we introduce how gut bacteria and different microbiota-derived metabolites, their receptors and signalling pathways can regulate the development of adipose tissue and its metabolic capacities. Finally, we describe the key challenges in moving from bench to bedside by presenting specific key examples.
Collapse
Affiliation(s)
- Patrice D Cani
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
18
|
Bernier-Latmani J, González-Loyola A, Petrova TV. Mechanisms and functions of intestinal vascular specialization. J Exp Med 2024; 221:e20222008. [PMID: 38051275 PMCID: PMC10697212 DOI: 10.1084/jem.20222008] [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: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
The intestinal vasculature has been studied for the last 100 years, and its essential role in absorbing and distributing ingested nutrients is well known. Recently, fascinating new insights into the organization, molecular mechanisms, and functions of intestinal vessels have emerged. These include maintenance of intestinal epithelial cell function, coping with microbiota-induced inflammatory pressure, recruiting gut-specific immune cells, and crosstalk with other organs. Intestinal function is also regulated at the systemic and cellular levels, such that the postprandial hyperemic response can direct up to 30% of systemic blood to gut vessels, while micron-sized endothelial cell fenestrations are necessary for nutrient uptake. In this review, we will highlight past discoveries made about intestinal vasculature in the context of new findings of molecular mechanisms underpinning gut function. Such comprehensive understanding of the system will pave the way to breakthroughs in nutrient uptake optimization, drug delivery efficiency, and treatment of human diseases.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | | | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Freire T, Hu Z, Wood KB, Gjini E. Modeling spatial evolution of multi-drug resistance under drug environmental gradients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567447. [PMID: 38014279 PMCID: PMC10680811 DOI: 10.1101/2023.11.16.567447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Multi-drug combinations to treat bacterial populations are at the forefront of approaches for infection control and prevention of antibiotic resistance. Although the evolution of antibiotic resistance has been theoretically studied with mathematical population dynamics models, extensions to spatial dynamics remain rare in the literature, including in particular spatial evolution of multi-drug resistance. In this study, we propose a reaction-diffusion system that describes the multi-drug evolution of bacteria, based on a rescaling approach (Gjini and Wood, 2021). We show how the resistance to drugs in space, and the consequent adaptation of growth rate is governed by a Price equation with diffusion. The covariance terms in this equation integrate features of drug interactions and collateral resistances or sensitivities to the drugs. We study spatial versions of the model where the distribution of drugs is homogeneous across space, and where the drugs vary environmentally in a piecewise-constant, linear and nonlinear manner. Applying concepts from perturbation theory and reaction-diffusion equations, we propose an analytical characterization of average mutant fitness in the spatial system based on the principal eigenvalue of our linear problem. This enables an accurate translation from drug spatial gradients and mutant antibiotic susceptibility traits, to the relative advantage of each mutant across the environment. Such a mathematical understanding allows to predict the precise outcomes of selection over space, ultimately from the fundamental balance between growth and movement traits, and their diversity in a population.
Collapse
Affiliation(s)
- Tomas Freire
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Zhijian Hu
- Departments of Biophysics and Physics, University of Michigan, USA
| | - Kevin B. Wood
- Departments of Biophysics and Physics, University of Michigan, USA
| | - Erida Gjini
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
20
|
Richard N, Savoye G, Leboutte M, Amamou A, Ghosh S, Marion-Letellier R. Crohn’s disease: Why the ileum? World J Gastroenterol 2023; 29:3222-3240. [PMID: 37377591 PMCID: PMC10292140 DOI: 10.3748/wjg.v29.i21.3222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/23/2023] [Accepted: 05/08/2023] [Indexed: 06/01/2023] Open
Abstract
Crohn’s disease (CD) is an inflammatory bowel disease characterized by immune-mediated flares affecting any region of the intestine alternating with remission periods. In CD, the ileum is frequently affected and about one third of patients presents with a pure ileal type. Moreover, the ileal type of CD presents epidemiological specificities like a younger age at onset and often a strong link with smoking and genetic susceptibility genes. Most of these genes are associated with Paneth cell dysfunction, a cell type found in the intestinal crypts of the ileum. Besides, a Western-type diet is associated in epidemiological studies with CD onset and increasing evidence shows that diet can modulate the composition of bile acids and gut microbiota, which in turn modulates the susceptibility of the ileum to inflammation. Thus, the interplay between environmental factors and the histological and anatomical features of the ileum is thought to explain the specific transcriptome profile observed in CD ileitis. Indeed, both immune response and cellular healing processes harbour differences between ileal and non-ileal CD. Taken together, these findings advocate for a dedicated therapeutic approach to managing ileal CD. Currently, interventional pharmacological studies have failed to clearly demonstrate distinct response profiles according to disease site. However, the high rate of stricturing disease in ileal CD requires the identification of new therapeutic targets to significantly change the natural history of this debilitating disease.
Collapse
Affiliation(s)
- Nicolas Richard
- University of Rouen Normandie, INSERM, ADEN UMR 1073, Nutrition, Inflammation and Microbiota-Gut-Brain Axis, Rouen F-76000, France
- CHU Rouen, Department of Gastroenterology, Rouen University Hospital-Charles Nicolle, Rouen F-76000, France
- Institute for Research and Innovation in Biomedicine, University of Rouen Normandie, Rouen F-76000, France
| | - Guillaume Savoye
- University of Rouen Normandie, INSERM, ADEN UMR 1073, Nutrition, Inflammation and Microbiota-Gut-Brain Axis, Rouen F-76000, France
- CHU Rouen, Department of Gastroenterology, Rouen University Hospital-Charles Nicolle, Rouen F-76000, France
- Institute for Research and Innovation in Biomedicine, University of Rouen Normandie, Rouen F-76000, France
| | - Mathilde Leboutte
- University of Rouen Normandie, INSERM, ADEN UMR 1073, Nutrition, Inflammation and Microbiota-Gut-Brain Axis, Rouen F-76000, France
- Institute for Research and Innovation in Biomedicine, University of Rouen Normandie, Rouen F-76000, France
| | - Asma Amamou
- APC Microbiome Ireland, Biosciences Building, University College Cork, Cork T12 YT20, Ireland
| | - Subrata Ghosh
- APC Microbiome Ireland, Biosciences Building, University College Cork, Cork T12 YT20, Ireland
| | - Rachel Marion-Letellier
- University of Rouen Normandie, INSERM, ADEN UMR 1073, Nutrition, Inflammation and Microbiota-Gut-Brain Axis, Rouen F-76000, France
- Institute for Research and Innovation in Biomedicine, University of Rouen Normandie, Rouen F-76000, France
| |
Collapse
|
21
|
Dal Co A, Ackermann M, van Vliet S. Spatial self-organization of metabolism in microbial systems: A matter of enzymes and chemicals. Cell Syst 2023; 14:98-108. [PMID: 36796335 DOI: 10.1016/j.cels.2022.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/14/2022] [Accepted: 12/21/2022] [Indexed: 02/17/2023]
Abstract
Most bacteria live in dense, spatially structured communities such as biofilms. The high density allows cells to alter the local microenvironment, whereas the limited mobility can cause species to become spatially organized. Together, these factors can spatially organize metabolic processes within microbial communities so that cells in different locations perform different metabolic reactions. The overall metabolic activity of a community depends both on how metabolic reactions are arranged in space and on how they are coupled, i.e., how cells in different regions exchange metabolites. Here, we review mechanisms that lead to the spatial organization of metabolic processes in microbial systems. We discuss factors that determine the length scales over which metabolic activities are arranged in space and highlight how the spatial organization of metabolic processes affects the ecology and evolution of microbial communities. Finally, we define key open questions that we believe should be the main focus of future research.
Collapse
Affiliation(s)
- Alma Dal Co
- Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland
| | - Martin Ackermann
- Department of Environmental Systems Science, ETH Zurich, 8092 Zurich, Switzerland; Department of Environmental Microbiology, Eawag, 8600 Duebendorf, Switzerland.
| | | |
Collapse
|
22
|
Liu J, Lu R, Zheng X, Hou W, Wu X, Zhao H, Wang G, Tian T. Establishment of a gut-on-a-chip device with controllable oxygen gradients to study the contribution of Bifidobacterium bifidum to inflammatory bowel disease. Biomater Sci 2023; 11:2504-2517. [PMID: 36779280 DOI: 10.1039/d2bm01490d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Supplemental Bifidobacterium has been shown to aid in the prevention, alleviation, and treatment of inflammatory bowel disease (IBD), but the progression and mechanisms are largely unstudied, partly because of a lack of appropriate models. In vitro human gut models must accurately recreate oxygen concentration gradients consistent with those in vivo to mimic gene expression, metabolism, and host-microbiome interactions. A non-equipment-intensive and inexpensive method for constructing the gut-on-a-chip with physiological oxygen concentration gradients remains challenging. Here, we propose a simple strategy using numerical simulations in a dual-channel gut-on-a-chip to guide chip design and achieve controllable oxygen gradients. By varying the size of microchannels, blocking the oxygen penetration of the polydimethylsiloxane layer at a given location, and controlling the flow of hypoxic/aerobic media, this strategy creates steep gradients across the intestinal epithelium. IBD symptoms were induced on the chip by tumor necrosis factor-α and lipopolysaccharide treatment. Bifidobacterium bifidum has been validated to contribute to the stability of the intestinal epithelial barrier, including preventing epithelial barrier disruption and promoting the repair of damaged intestinal epithelial cell monolayers. These effects may be associated with the co-localization of Bifidobacterium bifidum and ZO-1. This simple but robust approach for designing microfluidic devices is applicable to various organs-on-chips in which fluid dynamics and concentration profiles between different media must be considered. With the customized chip, the integration of activated Bifidobacterium bifidum provides an initial step toward developing a multi-factorial IBD platform. The approach could be scaled up for disease modeling, high-throughput drug screening and personalized medicine.
Collapse
Affiliation(s)
- Jun Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Ronghao Lu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Xiaolin Zheng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Wensheng Hou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Xiaoying Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Hezhao Zhao
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Tian Tian
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
23
|
Choo J, Glisovic N, Matic Vignjevic D. Gut homeostasis at a glance. J Cell Sci 2022; 135:281168. [DOI: 10.1242/jcs.260248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
ABSTRACT
The intestine, a rapidly self-renewing organ, is part of the gastrointestinal system. Its major roles are to absorb food-derived nutrients and water, process waste and act as a barrier against potentially harmful substances. Here, we will give a brief overview of the primary functions of the intestine, its structure and the luminal gradients along its length. We will discuss the dynamics of the intestinal epithelium, its turnover, and the maintenance of homeostasis. Finally, we will focus on the characteristics and functions of intestinal mesenchymal and immune cells. In this Cell Science at a Glance article and the accompanying poster, we aim to present the most recent information about gut cell biology and physiology, providing a resource for further exploration.
Collapse
Affiliation(s)
- Jieun Choo
- Institut Curie, PSL Research University, CNRS UMR 144 , F-75005 Paris , France
| | - Neda Glisovic
- Institut Curie, PSL Research University, CNRS UMR 144 , F-75005 Paris , France
| | | |
Collapse
|
24
|
Relationship between mucosa-associated gut microbiota and human diseases. Biochem Soc Trans 2022; 50:1225-1236. [PMID: 36214382 PMCID: PMC9704521 DOI: 10.1042/bst20201201] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022]
Abstract
The mucus layer covering the gastrointestinal (GI) tract plays a critical role in maintaining gut homeostasis. In the colon, the inner mucus layer ensures commensal microbes are kept at a safe distance from the epithelium while mucin glycans in the outer mucus layer provide microbes with nutrients and binding sites. Microbes residing in the mucus form part of the so-called 'mucosa-associated microbiota' (MAM), a microbial community which, due to its close proximity to the epithelium, has a profound impact on immune and metabolic health by directly impacting gut barrier function and the immune system. Alterations in GI microbial communities have been linked to human diseases. Although most of this knowledge is based on analysis of the faecal microbiota, a growing number of studies show that the MAM signature differs from faecal or luminal microbiota and has the potential to be used to distinguish between diseased and healthy status in well-studied conditions such as IBD, IBS and CRC. However, our knowledge about spatial microbial alterations in pathogenesis remains severely hampered by issues surrounding access to microbial communities in the human gut. In this review, we provide state-of-the-art information on how to access MAM in humans, the composition of MAM, and how changes in MAM relate to changes in human health and disease. A better understanding of interactions occurring at the mucosal surface is essential to advance our understanding of diseases affecting the GI tract and beyond.
Collapse
|
25
|
Livne N, Vaknin A. Collective responses of bacteria to a local source of conflicting effectors. Sci Rep 2022; 12:4928. [PMID: 35322063 PMCID: PMC8943191 DOI: 10.1038/s41598-022-08762-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/09/2022] [Indexed: 12/18/2022] Open
Abstract
To cope in complex environments, motile bacteria have developed a chemosensory system that integrates multiple cues and directs their motion toward regions that it deems favorable. However, we have a limited understanding of the principles that govern bacterial behavior in complex stimuli fields. Here, we followed the spatial redistribution of E. coli cells in perplexing environments created by a local source of both beneficial (nutrients) and hazardous (low pH or indole) effectors. We identified two fundamentally distinct collective responses: a ‘trade-off’ response, in which bacteria sharply accumulated at a distance from the source that reflected a trade-off between the propagating effectors, and a ‘bet-hedging’ response, in which part of the bacteria accumulated away from the source, avoiding the hazardous effector, while the other part evaded the repulsive force and accumulated at the source. In addition, we demonstrate that cells lacking the Tsr sensor swim toward both repellents and, surprisingly, even toward pH values well below 7. Using a numerical analysis, we could correlate the collective bacterial responses with fundamentally distinct chemotactic force fields created along the channel by the propagation of the effectors and their unique perception by the chemosensory system.
Collapse
Affiliation(s)
- Nir Livne
- The Racah Institute of Physics, The Hebrew University, Edmond J. Safra Campus, 91904, Jerusalem, Israel
| | - Ady Vaknin
- The Racah Institute of Physics, The Hebrew University, Edmond J. Safra Campus, 91904, Jerusalem, Israel.
| |
Collapse
|