Gut Microbiome–Brain Alliance: A Landscape View into Mental and Gastrointestinal Health and DisordersClick to copy article linkArticle link copied!
- Janet M. SassoJanet M. SassoCAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United StatesMore by Janet M. Sasso
- Ramy M. AmmarRamy M. AmmarBayer Consumer Health, R&D Digestive Health, Darmstadt 64295, GermanyMore by Ramy M. Ammar
- Rumiana TenchovRumiana TenchovCAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United StatesMore by Rumiana Tenchov
- Steven LemmelSteven LemmelCAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United StatesMore by Steven Lemmel
- Olaf Kelber
- Malte GrieswelleMalte GrieswelleBayer Consumer Health, R&D Digestive Health, Darmstadt 64295, GermanyMore by Malte Grieswelle
- Qiongqiong Angela Zhou*Qiongqiong Angela Zhou*Email: [email protected]CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United StatesMore by Qiongqiong Angela Zhou
Abstract
Gut microbiota includes a vast collection of microorganisms residing within the gastrointestinal tract. It is broadly recognized that the gut and brain are in constant bidirectional communication, of which gut microbiota and its metabolic production are a major component, and form the so-called gut microbiome–brain axis. Disturbances of microbiota homeostasis caused by imbalance in their functional composition and metabolic activities, known as dysbiosis, cause dysregulation of these pathways and trigger changes in the blood–brain barrier permeability, thereby causing pathological malfunctions, including neurological and functional gastrointestinal disorders. In turn, the brain can affect the structure and function of gut microbiota through the autonomic nervous system by regulating gut motility, intestinal transit and secretion, and gut permeability. Here, we examine data from the CAS Content Collection, the largest collection of published scientific information, and analyze the publication landscape of recent research. We review the advances in knowledge related to the human gut microbiome, its complexity and functionality, its communication with the central nervous system, and the effect of the gut microbiome–brain axis on mental and gut health. We discuss correlations between gut microbiota composition and various diseases, specifically gastrointestinal and mental disorders. We also explore gut microbiota metabolites with regard to their impact on the brain and gut function and associated diseases. Finally, we assess clinical applications of gut-microbiota-related substances and metabolites with their development pipelines. We hope this review can serve as a useful resource in understanding the current knowledge on this emerging field in an effort to further solving of the remaining challenges and fulfilling its potential.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Landscape of Gut Microbiome Research─Insights from the CAS Content Collection
Gut-Microbiota-Participant Bacteria
Techniques Used to Study the Gut Microbiome
Gastrointestinal Tract
Types of Bacteria Found in the GI Tract
Bacteroidetes
Firmicutes
Actinobacteria
Proteobacteria
Verrucomicrobia
Fusobacteria
Gut Microbiome–Disease Correlations
Digestive System Diseases and Disorders
Mental and Neurodegenerative Disorders
Metabolic Disorders
COVID-19
diseases | ↓ decreasing bacteria | ↑ increasing bacteria |
---|---|---|
digestive system diseases | ||
irritable bowel syndrome (194−199) | Bifidobacterium | Ruminococcus |
Faecalibacterium prausnitzii | Dorea | |
Bacteroides | Enterobacteriaceae | |
Lactobacillaceae | ||
Bacteroides | ||
Firmicutes/Bacteroidetes ratio | ||
IBD: Crohn’s disease (200,201) | Bacteroides | |
Faecalibacterium prausnitzii | ||
Bifidobacterium adolescentis | ||
IBD: ulcerative colitis (139,200) | Bifidobacteria | |
Roseburia hominis | ||
Faecalibacterium prausnitzii | ||
Lachnospiraceae | ||
Ruminococcaceae | ||
mental health disorders | ||
anxiety disorder (202,203) | Bacteriodetes | Bacteroidaceae |
Ruminococcus gnavus | Enterobacteriaceae | |
Fusobacterium | Burkholderiaceae | |
post-traumatic stress disorder (204) | Actinobacteria | |
Lentisphaerae | ||
Verrucomicrobia | ||
depression (205−207) | Prevotella | Eggerthella |
Dialister | Holdemania | |
Turicibacter | ||
Paraprevotella | ||
dementia (172) | Actinobacteria | Escherichia |
Bacteroides | Blautia | |
Bifidobacterium | ||
Streptococcus | ||
Lactobacillus | ||
Dorea | ||
metabolic disorders | ||
diabetes type 1 (208,209) | Lactobacillus | Clostridium |
Bifidobacterium | Bacteroides | |
Blautia coccoides | Veillonella | |
Eubacterium rectale | Actinobacteria | |
Prevotella | Proteobacteria | |
Akkermansia | Lactococcus | |
Firmicutes | ||
diabetes type 2 (199,209,210) | Firmicutes | Betaproteobacteria |
Clostridia | ||
Lactobacillus | Bacteroidetes/Firmicutes ratio | |
Akkermansia muciniphilia | ||
Roseburia | ||
obesity (199,211) | Bacteroidetes | Enterobacteria |
Methanobrevibacter smithii | Ruminococcus gnavus | |
Ruminococcus flavefaciens | Actinobacteria | |
Bifidobacterium | Prevotellaceae |
Gut Bacteria–Disease Correlations
Therapeutic Strategies for the Treatment of Mental and Gastrointestinal Disorders
Dietary Interventions
Probiotics and Prebiotics
Antibiotics
Fecal Microbiota Transplantation (FMT)
Psychotherapeutic Interventions
Pharmacological Interventions
Gut Microbiota Metabolites
Gut Microbiota Metabolites with Impact on Brain Function
metabolite class/references | specific functions | associated diseases |
---|---|---|
short-chain fatty acids (258,285−290) | – gut microbiota composition regulation | – diabetes |
– gut barrier integrity support | – obesity | |
– energy homeostasis support | – nonalcoholic fatty liver disease | |
– gut hormone production | – ulcerative colitis | |
– circadian rhythm regulation | – Crohn’s disease | |
– proinflammatory cytokines inhibition | – colorectal cancer | |
– immunomodulation | – autism spectrum disorder | |
– water, sodium, calcium, magnesium absorption | – Parkinson’s disease | |
– regulation of intestinal pH value | – diarrhea | |
– IBS | ||
– constipation | ||
– functional dyspepsia (FD) | ||
bile acids (BAs) (291−295) | – lipid and vitamin absorption regulation | – obesity |
– gut microbiota composition regulation | – nonalcoholic steatohepatitis | |
– gut hormones production | – ulcerative colitis | |
– intestinal immunity | – cancer | |
– intestinal electrolyte and fluid balance | – multiple sclerosis | |
– gut motility | – Alzheimer’s disease | |
– gut barrier integrity | – Parkinson’s disease | |
– lipid homeostasis | – traumatic brain injury | |
– glucose homeostasis | – stroke | |
– amino acid homeostasis | – amyotrophic lateral sclerosis | |
– circadian rhythm | – IBS | |
– neurotransmission | ||
tryptophan and indole derivatives (296−300) | – gut microbial spore formation | – ulcerative colitis |
– drug resistance | – Crohn’s disease | |
– biofilm formation | – obesity | |
– intestinal barrier function regulation | – stroke | |
– gut hormone secretion | – mucosal candidiasis | |
– gut motility | – autism spectrum disorder | |
– immunomodulation | – Alzheimer’s disease | |
– Parkinson’s disease | ||
– migraine | ||
– schizophrenia | ||
– IBS | ||
choline metabolites (301−303) | – bile acid synthesis inhibition | – nonalcoholic fatty liver disease |
– inflammation promotion | – obesity | |
– thrombosis | – diabetes | |
– myocardial hypertrophy and fibrosis | – hypertension | |
– mitochondrial dysfunction exacerbation | ||
vitamins (304−306) | – cellular metabolism regulation | – vitamin-associated diseases |
– immunomodulation | – schizophrenia | |
– cell proliferation | – autism | |
– vitamins supply | – dementia | |
– IBS | ||
– IBD | ||
neurotransmitters (307−309) | – gut motility regulation | – Parkinson’s disease |
– memory support | – autism spectrum disorder | |
– stress response | – IBD | |
– nervous system | – IBS | |
– immune response | ||
lipids (184,310,311) | – systemic inflammation promotion | – diabetes |
– hyperinsulinemia regulation | – obesity | |
– immunomodulation | – nonalcoholic fatty liver disease | |
– bile acid synthesis | – hyperinsulinemia | |
– hypercholesterolemia | ||
– chronic hepatitis C | ||
gases (307,312−316) | – gut motility | – colitis |
– gut inflammation | – ulcer | |
– epithelial secretion | – IBS | |
– mucosal blood flow |
Gut Microbiota Metabolites’ Role in Digestive System
Gut Microbiome–Brain Axis
Irritable Bowel Syndrome
Functional Dyspepsia
Functional Constipation
Stress and Stress Resilience
Sleep
Cognitive Function
Emotional Well-being
Prebiotics, Probiotics, Synbiotics, Postbiotics, and Psychobiotics
Probiotics
Prebiotics
Synbiotics
Postbiotics
Psychobiotics
Pre-, Pro-, Postbiotics, and Fecal Microbiota Transplantation in the Development Pipelines
Private Investment
Companies and Academic Institutions Investigating Treatment of Mental Disorders and DGBI through Gut Microbiome Modulation
Probiotics
Prebiotics
Postbiotics
Fecal Microbiota Transplantation
Clinical Trials Landscape for Probiotics in Mental Disorders and DGBI
clinical trial identifier | condition | intervention | status |
---|---|---|---|
NCT05564767 (472) | depression, anxiety, stress | Bifidobacterium adolescentis Bif-038, Lacticaseibacillus rhamnosus LGG, Bifidobacterium BB-12 | recruiting |
NCT03494725 (477) | stress, anxiety | Lacticaseibacillus paracasei Lpc-37 | complete |
NCT04767997 (474) | sleep disorder | undisclosed probiotic formulation | recruiting |
NCT03601559 (478) | cognitive impairment | Lactobacillus paracasei Lpc-37 | complete |
NCT03615651 (479) | stress, cognition impairment | Lactobacillus helveticus, Bifidobacterium longum, Lactiplantibacillus plantarum | complete |
NCT05567653 (473) | stress | Lactobacillus helveticus Rosell-52, Bifidobacterium longum Rosell-175 | recruiting |
NCT03370458 (480) | stress | Lactobacillus plantarum DR7 | complete |
clinical trial identifier | condition | probiotic intervention | status |
---|---|---|---|
NCT02592200 (485) | functional constipation | Lactobacillus gasseri DSM 27123 | complete |
NCT04304170 (486) | functional constipation | Bifidobacterium animalis lactis (LMG P-28145) | complete |
NCT04662957 (487) | diarrhea-predominant irritable bowel syndrome | Bifidobacterium breve BB010, Bifidobacterium longum BL020, Bifidobacterium bifidum BF030, Bifidobacterium lactis BL040, Lactobacillus rhamnosus LR110, Lactobacillus paracasei LPC100, Lactobacillus acidophilus LA120, Lactobacillus casei LC130, Lactobacillus plantarum LP140, Streptococcus thermophilus ST25 | complete |
NCT05566171 (488) | functional constipation | Bifidobacterium lactis CNCM I-2494, Bifidobacterium lactis DN 173-010 | enrolling by invitation |
NCT01463293 (489) | functional constipation | Bifidobacterium lactis HN019 | complete |
NCT01102036 (490) | functional constipation | Lactobacillus paracasei F19, Lactobacillus paracasei LA-5, Bifidobacterium lactis BB-12 | complete |
NCT03721107 (491) | diarrhea- and constipation-predominant irritable bowel syndrome | Blautia hydrogenotrophica | complete |
NCT00534170 (492) | functional diarrhea | Lactobacillus casei Shirota, Bifidobacterium breve Yakult | complete |
NCT00794924 (493) | functional diarrhea, functional constipation | Streptococcus thermophilus, Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus delbrueckii subsp. bulgaricus | complete |
NCT02213172 (494) | irritable bowel syndrome | Bifidobacterium longum R0175, Lactobacillus paracasei HA-196 | complete |
NCT00807326 (495) | functional diarrhea | Saccharomyces boulardii | complete |
NCT05054309 (496) | irritable bowel syndrome | Bifidobacterium longum NCC3001 | recruiting |
NCT01099696 (497) | functional dyspepsia | Bifidobacterium infantis 35624 | complete |
NCT01887834 (498) | irritable bowel syndrome | Lactobacillus gasseri, Bifidobacterium bifidum, Bifidobacterium longum | complete |
NCT04605783 (499) | functional diarrhea | Saccharomyces boulardii CNCM I-745 | not yet recruiting |
NCT04950296 (500) | irritable bowel syndrome with diarrhea | Lactobacillus plantarum UALp-05 | complete |
NCT05149599 (501) | irritable bowel syndrome | Saccharomyces cerevisiae | complete |
Mental Disorders
DGBI
Clinical Trials Landscape for Prebiotics in Mental Disorders and DGBI
clinical trial identifier | condition | intervention | status |
---|---|---|---|
NCT05372601 (504) | stress | GOS | complete |
NCT05239845 (505) | sleep disorder | polydextrose, GOS | recruiting |
NCT04324749 (506) | cognitive impairment, stress | roasted peanuts, peanut butter | complete |
NCT05528575 (507) | stress | GOS, inulin, resistant potato starch RS2 | active |
NCT04616937 (508) | anxiety, cognitive impairment | GOS | complete |
clinical trial identifier | condition | intervention | status |
---|---|---|---|
ISRCTN54052375 (512) | irritable bowel syndrome | GOS | complete |
NCT04491734 (513) | gastresophageal reflux | maltosyl-isomaltooligosaccharides (MIMO) | complete |
NCT05207618 (514) | irritable bowel syndrome with diarrhea | chestnut and quebracho tannin extract | complete |
ACTRN12612001270808 (510) | functional constipation | green kiwi prebiotic, gold kiwi prebiotic | complete |
NCT05340712 (515) | functional constipation | infant formula with lactose (prebiotic) along with probiotics | recruiting |
Mental Disorders
DGBI
Clinical Trials Landscape for Postbiotics and FMT in Mental Disorders and DGBI
clinical trial identifier | condition | intervention | status |
---|---|---|---|
NCT05475314 (517) | irritable bowel syndrome | microbially fermented postbiotic oat drink | complete |
NCT05562739 (518) | anxiety | multistrain postbiotic | not yet recruiting |
NCT05339243 (519) | irritable bowel syndrome with diarrhea | heat-treated Bifidobacterium longum ES1 | recruiting |
clinical trial identifier | condition | intervention | status |
---|---|---|---|
NCT03822299 (522) | irritable bowel syndrome | fecal microbiota transplantation | complete |
NCT02092402 (523) | irritable bowel syndrome | fecal microbiota transplantation | complete |
NCT05035784 (524) | functional constipation | fecal microbiota transplantation | recruiting |
NCT05427331 (525) | chronic insomnia | fecal microbiota transplantation capsule | recruiting |
Postbiotic Clinical Trials Landscape for the Treatment of Mental Disorders and DGBI
FMT Clinical Trials Landscape for the Treatment of Mental Disorders and DGBI
Noteworthy Probiotic and Prebiotic Patents
patent number | title | summary |
---|---|---|
WO2016085356A1 | gold kiwifruit compositions and methods of preparation and use thereof | Prebiotic compositions prepared from gold varieties of Actinidia chinensis. These prebiotic compositions treat or prevent DGBI, such as constipation, and IBS. |
WO2022191767A1 | GOS preconditioning L. reuteri and GOS in final formulation | Enhancing the survival and activity of probiotic Lactobacillus reuteri strains by preconditioning L. reuteri with GOS. This method produces high synbiotic and beneficial effects of the probiotic bacteria in the gastrointestinal tract, such as boosting calcium and iron solubility, along with enhancing the production of lactic and acetic acid. |
US20160058808A1 | microbe-based modulation of serotonin biosynthesis | Methods and probiotic compositions that can be used to modulate serotonin levels and adjust the composition of gut microbiota along with adjusting the level of serotonin-related metabolites. |
WO2022182908A1 | probiotic therapies for social deficit and stress response | Bacterial species, including probiotic Enterococcus faecalis, for use in the treatment of social behavioral deficit symptoms, such as depression, by increasing social behavior and decreasing corticosterone levels along with c-Fos expression in the brain. |
US9192618B2 | method of treating constipation-predominant irritable bowel syndrome | Prebiotic or probiotic agent that inhibits the growth of methanogenic bacteria or promotes the growth of competing intestinal microbiotia for the treatment of constipation predominant IBS. |
US10022408B2 | probiotic Bifidobacterium adolescentis strains | Novel isolated strains of probiotic Bifidobacterium adolescentis for the prevention, alleviation of symptoms, or treatment of intestinal inflammatory conditions, such as IBS. |
WO2005003329A1 | novel GOS composition and the preparation thereof | Novel strains of Bifidobacterium hifidum capable of producing a novel galactosidase enzyme activity that converts lactose to a novel mixture of GOS. The mixture of prebiotic oligosaccharides improves gut health by promoting the growth of bifidobacteria in the gut. |
US20220040242A1 | modulation of the gut microbiome to treat mental disorders or diseases of the central nervous system | Methods of treating at least one symptom of a mental disorder and the central nervous system by modulating the amount of GABA produced in the gut. Also disclosed are methods of identifying and creating probiotic bacterial strains capable of producing GABA. |
WO2016029198A1 | process for the production of isomaltooligosaccharide | Provides a method for the production of prebiotic oligosaccharides by the fermentation of dextransucrase-producing microorganisms. |
WO2022214700A1 | Lacticaseibacillus paracasei EM025-11 and uses thereof | A probiotic strain of Lacticaseibacillus paracasei EM025-11 that adheres to intestinal epithelial cells and has anti-inflammatory activity by upregulating genes associated with immune engagement for the treatment of IBS with constipation. |
US20220280576A1 | Bifidobacterium longum and functional GI disorders | Methods for treating functional GI disorders with probiotic Bifidobacterium longum ATCC BAA-999. |
WO2019149941A1 | postbiotic-based composition for the modulation of immune system activation and protection of mucosal barriers | A fermented supernatant of Lactobacillus casei or paracasei species for the promotion of human health and prevention of inflammatory disorders. The postbiotic was shown to stimulate peripheral blood mononuclear cells and protect from endotoxic shock and Salmonella infection. |
US8551498B2 | solid composition containing Bacillus-type nonpathogenic bacterial spores | Composition of spores of probiotic bacteria Bacillus useful in the pharmaceutical, veterinary, and nutrition fields. |
ES2824536T3 | use of microbial communities for human and animal health | Mixture of probiotic bacteria belonging to at least six or seven bacterial species to prevent or treat GI disorders. |
US20220233559A1 | xylooligosaccharide as a multifunctional prebiotic | Prebiotic mixture of xylooligosaccharides derived from sugar cane that were shown to modulate the levels of probiotic bacteria Bifidobacteria and Lactobacillus in the microbiome. |
WO2022173764A1 | nutritional plant-based foods and beverages, methods of manufacture, and methods of treatment | Formulation of a prebiotic beverage whose ingestion modulates the gut microbiome by enhancing the growth of Bifidobacterium, improving the production of SCFAs, and reducing the levels Escherichia coli. |
US20220315960A1 | method for producing gamma-aminobutyric acid and fermented culture prepared thereby | Process to produce gamma-aminobutyric acid from glutamic acid and a probiotic composition capable of this biotransformation. Ideally, the composition contains different probiotic bacteria Bifidobacterium and Lactobacillus strains. |
WO2022208458A1 | inactivated strains of bacteria, such as viable but nonculturable bacteria, compositions, and use thereof | Postbiotic composition of different gamma-irradiated members of bacterial species Lactobacillus, Lacticaseibacillus, Bifidobacterium, and Lactiplantibacillus to treat several gastrointestinal disorders. |
EP3932415A1 | gut microbiota composition and uses thereof | Probiotic composition for the prevention and/or treatment of a mental disorder with memory impairment by gut microbiome modulation for an increase in memory scores. |
Conclusions and Perspective
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschemneuro.3c00127.
Prebiotic, probiotic, postbiotic, and fecal transplant therapeutic clinical trial data investigating the treatment of mental disorders and DGBI through gut microbiome modulation (XLSX)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
The authors sincerely appreciate the CAS Data, Analytics & Insights team for their assistance in data extraction. The authors also appreciate Dharmini Patel for project coordination, Manuela Pausan for ideation, and Peter Jap and Cristina Tomeo for insightful discussion. The authors are also grateful to Manuel Guzman, Gilles Georges, Michael Dennis, Dawn Riedel, Dawn George, and Hong Xie for executive sponsorship.
4-EPS | 4-ethylphenylsulfate |
5-AVAB | 5-aminovaleric acid betaine |
BAs | bile acids |
BBB | blood-brain barrier |
BCFA | branched-chain fatty acids |
CAN | central autonomic network |
CEN | central executive network |
CNS | central nervous system |
DGBI | disorders of gut–brain interaction |
DMN | default mode network |
EAN | emotional arousal network |
ENS | enteric nervous system |
FC | functional constipation |
FCT | fecal microbiota transplantation |
FD | functional dyspepsia |
FGIDs | functional gastrointestinal disorders |
GBA | gut–brain axis |
HC | healthy controls |
HPA | hypothalamus–pituitary–adrenal |
IBS | irritable bowel syndrome |
IHMC | International Human Microbiome Consortium |
MPs | muramyl peptides |
OCC | occipital network |
RDA | recommended daily allowance |
SAL | salience network |
SCFA | short-chain fatty acids |
SMN | sensorimotor network |
TMAO | trimethylamine-N-oxide |
References
This article references 536 other publications.
- 1Schopf, J. W. Disparate rates, differing fates: tempo and mode of evolution changed from the Precambrian to the Phanerozoic. Proc. Natl. Acad. Sci. U. S. A. 1994, 91, 6735– 6742, DOI: 10.1073/pnas.91.15.6735Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXlslGjtr4%253D&md5=31cf08a8c7c02f915feadc74ded5ef7bDisparate rates, differing fates: tempo and mode of evolution changed from the Precambrian to the PhanerozoicSchopf, J. WilliamProceedings of the National Academy of Sciences of the United States of America (1994), 91 (15), 6735-42CODEN: PNASA6; ISSN:0027-8424.Over the past quarter century, detailed genus- and species-level similarities in cellular morphol. between described taxa of Precambrian microfossils and extant cyanobacteria have been noted and regarded as biol. and taxonomically significant by numerous workers worldwide. Such similarities are particularly well documented for members of the Oscillatoriaceae and Chroococcaceae, the two most abundant and widespread Precambrian cyanobacterial families. For species of two addnl. families, the Entophysalidaceae and Pleurocapsaceae, species-level morphol. similarities are supported by in-depth fossil-modern comparisons of environment, taphonomy, development, and behavior. Morphol. and probably physiol. as well, such cyanobacterial "living fossils" have exhibited an extraordinarily slow (hypobradytelic) rate of evolutionary change, evidently a result of the broad ecol. tolerance characteristic of many members of the group and a striking example of G G. Simpson's [Simpson, G. G. (1944) Tempo and Mode in Evolution (Columbia Univ. Press, New York)] "rule of the survival of the relatively unspecialized.". In both tempo and mode of evolution, much of the Precambrian history of life-that dominated by microscopic cyanobacteria and related prokaryotes-appears to have differed markedly from the more recent Phanerozoic evolution of megascopic, horotelic, adaptationally specialized eukaryotes.
- 2Averill, C.; Anthony, M. A.; Baldrian, P.; Finkbeiner, F.; van den Hoogen, J.; Kiers, T.; Kohout, P.; Hirt, E.; Smith, G. R.; Crowther, T. W. Defending Earth’s terrestrial microbiome. Nat. Microbiol. 2022, 7, 1717, DOI: 10.1038/s41564-022-01228-3Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XisFCgurjP&md5=6f52ece9458ac70645ac49ffb979f409Defending Earth's terrestrial microbiomeAverill, Colin; Anthony, Mark A.; Baldrian, Petr; Finkbeiner, Felix; van den Hoogen, Johan; Kiers, Toby; Kohout, Petr; Hirt, Eliane; Smith, Gabriel Reuben; Crowther, Tom W.Nature Microbiology (2022), 7 (11), 1717-1725CODEN: NMAICH; ISSN:2058-5276. (Nature Portfolio)Abstr.: Microbial life represents the majority of Earth's biodiversity. Across disparate disciplines from medicine to forestry, scientists continue to discover how the microbiome drives essential, macro-scale processes in plants, animals and entire ecosystems. Yet, there is an emerging realization that Earth's microbial biodiversity is under threat. Here we advocate for the conservation and restoration of soil microbial life, as well as active incorporation of microbial biodiversity into managed food and forest landscapes, with an emphasis on soil fungi. We analyze 80 expts. to show that native soil microbiome restoration can accelerate plant biomass prodn. by 64% on av., across ecosystems. Enormous potential also exists within managed landscapes, as agriculture and forestry are the dominant uses of land on Earth. Along with improving and stabilizing yields, enhancing microbial biodiversity in managed landscapes is a crit. and underappreciated opportunity to build reservoirs, rather than deserts, of microbial life across our planet. As markets emerge to engineer the ecosystem microbiome, we can avert the mistakes of aboveground ecosystem management and avoid microbial monocultures of single high-performing microbial strains, which can exacerbate ecosystem vulnerability to pathogens and extreme events. Harnessing the planet's breadth of microbial life has the potential to transform ecosystem management, but it requires that we understand how to monitor and conserve the Earth's microbiome.
- 3O’Hara, A. M.; Shanahan, F. The gut flora as a forgotten organ. EMBO reports 2006, 7, 688– 693, DOI: 10.1038/sj.embor.7400731Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XmsVanurg%253D&md5=4976fe244704b626436ac3ac00f28c99The gut flora as a forgotten organO'Hara, Ann M.; Shanahan, FergusEMBO Reports (2006), 7 (7), 688-693CODEN: ERMEAX; ISSN:1469-221X. (Nature Publishing Group)A review. The intestinal microflora is a pos. health asset that crucially influences the normal structural and functional development of the mucosal immune system. Mucosal immune responses to resident intestinal microflora require precise control and an immunosensory capacity for distinguishing commensal from pathogenic bacteria. In genetically susceptible individuals, some components of the flora can become a liability and contribute to the pathogenesis of various intestinal disorders, including inflammatory bowel diseases. It follows that manipulation of the flora to enhance the beneficial components represents a promising therapeutic strategy. The flora has a collective metabolic activity equal to a virtual organ within an organ, and the mechanisms underlying the conditioning influence of the bacteria on mucosal homeostasis and immune responses are beginning to be unravelled. An improved understanding of this hidden organ will reveal secrets that are relevant to human health and to several infectious, inflammatory and neoplastic disease processes.
- 4Bäckhed, F.; Ley, R. E.; Sonnenburg, J. L.; Peterson, D. A.; Gordon, J. I. Host-bacterial mutualism in the human intestine. Science 2005, 307, 1915– 1920, DOI: 10.1126/science.1104816Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2M7lvVKlsg%253D%253D&md5=3f7557c5ea4d355c758e9bce97f544e9Host-bacterial mutualism in the human intestineBackhed Fredrik; Ley Ruth E; Sonnenburg Justin L; Peterson Daniel A; Gordon Jeffrey IScience (New York, N.Y.) (2005), 307 (5717), 1915-20 ISSN:.The distal human intestine represents an anaerobic bioreactor programmed with an enormous population of bacteria, dominated by relatively few divisions that are highly diverse at the strain/subspecies level. This microbiota and its collective genomes (microbiome) provide us with genetic and metabolic attributes we have not been required to evolve on our own, including the ability to harvest otherwise inaccessible nutrients. New studies are revealing how the gut microbiota has coevolved with us and how it manipulates and complements our biology in ways that are mutually beneficial. We are also starting to understand how certain keystone members of the microbiota operate to maintain the stability and functional adaptability of this microbial organ.
- 5Cénit, M. C.; Matzaraki, V.; Tigchelaar, E. F.; Zhernakova, A. Rapidly expanding knowledge on the role of the gut microbiome in health and disease. Biochim. Biophys. Acta 2014, 1842, 1981– 1992, DOI: 10.1016/j.bbadis.2014.05.023Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXpvVart74%253D&md5=46e82a3c41ac32b0a78c4c253b8365d9Rapidly expanding knowledge on the role of the gut microbiome in health and diseaseCenit, M. C.; Matzaraki, V.; Tigchelaar, E. F.; Zhernakova, A.Biochimica et Biophysica Acta, Molecular Basis of Disease (2014), 1842 (10), 1981-1992CODEN: BBADEX; ISSN:0925-4439. (Elsevier B. V.)A review. The human gut is colonized by a wide diversity of micro-organisms, which are now known to play a key role in the human host by regulating metabolic functions and immune homeostasis. Many studies have indicated that the genomes of the authors' gut microbiota, known as the gut microbiome or the authors' "other genome" could play an important role in immune-related, complex diseases, and growing evidence supports a causal role for gut microbiota in regulating predisposition to diseases. A comprehensive anal. of the human gut microbiome is thus important to unravel the exact mechanisms by which the gut microbiota are involved in health and disease. Recent advances in next-generation sequencing technol., along with the development of metagenomics and bioinformatics tools, have provided opportunities to characterize the microbial communities. Furthermore, studies using germ-free animals have shed light on how the gut microbiota are involved in autoimmunity. In this review the authors describe the different approaches used to characterize the human microbiome, review current knowledge about the gut microbiome, and discuss the role of gut microbiota in immune homeostasis and autoimmunity. Finally, the authors indicate how this knowledge could be used to improve human health by manipulating the gut microbiota. This article is part of a Special Issue entitled: From Genome to Function.
- 6Vrancken, G.; Gregory, A. C.; Huys, G. R. B.; Faust, K.; Raes, J. Synthetic ecology of the human gut microbiota. Nature Reviews Microbiology 2019, 17, 754– 763, DOI: 10.1038/s41579-019-0264-8Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvV2ktrbF&md5=d93899ca06271b82c58a83b2bfa455a9Synthetic ecology of the human gut microbiotaVrancken, Gino; Gregory, Ann C.; Huys, Geert R. B.; Faust, Karoline; Raes, JeroenNature Reviews Microbiology (2019), 17 (12), 754-763CODEN: NRMACK; ISSN:1740-1526. (Nature Research)Despite recent advances in sequencing and culturing, a deep knowledge of the wiring and functioning of the human gut ecosystem and its microbiota as a community is still missing. A holistic mechanistic understanding will require study of the gut microbiota as an interactive and spatially organized biol. system, which is difficult to do in complex natural communities. Synthetic gut microbial ecosystems can function as model systems to further current understanding of the compn., stability and functional activities of the microbiota. In this Review, we provide an overview of the current synthetic ecol. strategies that can be used towards a more comprehensive understanding of the human gut ecosystem. Such approaches that integrate in vitro expts. using cultured isolates with math. modeling will enable the ultimate goal: translating mechanistic and ecol. knowledge into novel and effective therapies.
- 7Hasan, N.; Yang, H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ. 2019, 7, e7502, DOI: 10.7717/peerj.7502Google ScholarThere is no corresponding record for this reference.
- 8Pariente, N.; York, A. Milestones in human microbiota research. https://www.nature.com/immersive/d42859-019-00041-z/index.html (accessed October 6, 2022).Google ScholarThere is no corresponding record for this reference.
- 9Malla, M. A.; Dubey, A.; Kumar, A.; Yadav, S.; Hashem, A.; Abd_Allah, E. F. Exploring the Human Microbiome: The Potential Future Role of Next-Generation Sequencing in Disease Diagnosis and Treatment. Front. Immunol. 2019, 9, 02868, DOI: 10.3389/fimmu.2018.02868Google ScholarThere is no corresponding record for this reference.
- 10Berg, G.; Rybakova, D.; Fischer, D.; Cernava, T.; Vergès, M.-C. C.; Charles, T.; Chen, X.; Cocolin, L.; Eversole, K.; Corral, G. H. Microbiome definition re-visited: old concepts and new challenges. Microbiome 2020, 8, 103, DOI: 10.1186/s40168-020-00875-0Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38nms1Sgug%253D%253D&md5=834e4c5e1470b19c81ccbbaf181b37d1Microbiome definition re-visited: old concepts and new challengesBerg Gabriele; Rybakova Daria; Cernava Tomislav; Fischer Doreen; Schloter Michael; Verges Marie-Christine Champomier; Maguin Emmanuelle; Charles Trevor; Charles Trevor; Chen Xiaoyulong; Cocolin Luca; Schelkle Bettina; Eversole Kellye; Corral Gema Herrero; Kazou Maria; Kinkel Linda; Lange Lene; Lima Nelson; Loy Alexander; Wagner Michael; Macklin James A; Mauchline Tim; McClure Ryan; Mitter Birgit; Sessitsch Angela; Ryan Matthew; Sarand Inga; Smidt Hauke; van Overbeek Leo; Roume Hugo; Kiran G Seghal; Selvin Joseph; Souza Rafael Soares Correa de; Singh Brajesh K; Singh Brajesh K; Walsh AaronMicrobiome (2020), 8 (1), 103 ISSN:.The field of microbiome research has evolved rapidly over the past few decades and has become a topic of great scientific and public interest. As a result of this rapid growth in interest covering different fields, we are lacking a clear commonly agreed definition of the term "microbiome." Moreover, a consensus on best practices in microbiome research is missing. Recently, a panel of international experts discussed the current gaps in the frame of the European-funded MicrobiomeSupport project. The meeting brought together about 40 leaders from diverse microbiome areas, while more than a hundred experts from all over the world took part in an online survey accompanying the workshop. This article excerpts the outcomes of the workshop and the corresponding online survey embedded in a short historical introduction and future outlook. We propose a definition of microbiome based on the compact, clear, and comprehensive description of the term provided by Whipps et al. in 1988, amended with a set of novel recommendations considering the latest technological developments and research findings. We clearly separate the terms microbiome and microbiota and provide a comprehensive discussion considering the composition of microbiota, the heterogeneity and dynamics of microbiomes in time and space, the stability and resilience of microbial networks, the definition of core microbiomes, and functionally relevant keystone species as well as co-evolutionary principles of microbe-host and inter-species interactions within the microbiome. These broad definitions together with the suggested unifying concepts will help to improve standardization of microbiome studies in the future, and could be the starting point for an integrated assessment of data resulting in a more rapid transfer of knowledge from basic science into practice. Furthermore, microbiome standards are important for solving new challenges associated with anthropogenic-driven changes in the field of planetary health, for which the understanding of microbiomes might play a key role. Video Abstract.
- 11Butler, M. I.; Mörkl, S.; Sandhu, K. V.; Cryan, J. F.; Dinan, T. G. The Gut Microbiome and Mental Health: What Should We Tell Our Patients?: Le microbiote Intestinal et la Santé Mentale: que Devrions-Nous dire à nos Patients?. Can. J. Psychiatry. 2019, 64, 747– 760, DOI: 10.1177/0706743719874168Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3Mrps1KlsA%253D%253D&md5=bd5b0b7edcb3e31a64274c036a7bee18The Gut Microbiome and Mental Health: What Should We Tell Our Patients?: Le microbiote Intestinal et la Sante Mentale : que Devrions-Nous dire a nos Patients?Butler Mary I; Morkl Sabrina; Dinan Timothy G; Morkl Sabrina; Sandhu Kiran V; Cryan John FCanadian journal of psychiatry. Revue canadienne de psychiatrie (2019), 64 (11), 747-760 ISSN:.The gut microbiome as a potential therapeutic target for mental illness is a hot topic in psychiatry. Trillions of bacteria reside in the human gut and have been shown to play a crucial role in gut-brain communication through an influence on neural, immune, and endocrine pathways. Patients with various psychiatric disorders including depression, bipolar disorder, schizophrenia, and autism spectrum disorder have been shown to have significant differences in the composition of their gut microbiome. Enhancing beneficial bacteria in the gut, for example, through the use of probiotics, prebiotics, or dietary change, has the potential to improve mood and reduce anxiety in both healthy people and patient groups. Much attention is being given to this subject in the general media, and patients are becoming increasingly interested in the potential to treat mental illness with microbiome-based therapies. It is imperative that those working with people with mental illness are aware of the rationale and current evidence base for such treatment strategies. In this review, we provide an overview of the gut microbiome, what it is, and what it does in relation to gut-brain communication and psychological function. We describe the fundamental principles and basic techniques used in microbiome-gut-brain axis research in an accessible way for a clinician audience. We summarize the current evidence in relation to microbiome-based strategies for various psychiatric disorders and provide some practical advice that can be given to patients seeking to try a probiotic for mental health benefit.
- 12Corliss, J. O. Three Centuries of Protozoology: A Brief Tribute to its Founding Father, A. van Leeuwenhoek of Delft. Journal of Protozoology 1975, 22, 3– 7, DOI: 10.1111/j.1550-7408.1975.tb00934.xGoogle Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE2M7hs1GhtQ%253D%253D&md5=879a6323e91fa81ac855aad29b9a525bThree centuries of protozoology: a brief tribute to its founding father, A. van Leeuwenhoek of DelftCorliss J OThe Journal of protozoology (1975), 22 (1), 3-7 ISSN:0022-3921.It was exactly 300 years ago this month (August 1974) that the 17th century modest draper from Delft, Holland--Antony van Leeuwenhoek--discovered protozoa. Describing them, often with amazing accuracy considering the optical equipment he was using (simply a home-made "glorified" hand lens), in letters to the Royal Society of London, he established himself, certainly, as the founding father of protozoology. It is particularly appropriate for an assemblage of protozoologists to pay homage to this intrepid "philosopher in little things," a man with an insatiable curiosity about his wee animalcules, on the tricentenary of his discovery of them, since it was an event of such long-lasting significance.
- 13Leidy, J. A flora and fauna within living animals. Smithsonian Institution: WA, 1853.Google ScholarThere is no corresponding record for this reference.
- 14Savage, D. C. Microbial biota of the human intestine: a tribute to some pioneering scientists. Curr. Issues Intest. Microbiol. 2001, 2, 1– 15Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3MnmsVOlsw%253D%253D&md5=75a3c7bab223cbc390e38f4b8d9644daMicrobial biota of the human intestine: a tribute to some pioneering scientistsSavage D CCurrent issues in intestinal microbiology (2001), 2 (1), 1-15 ISSN:1466-531X.Research on the indigenous intestinal microbiota of man was initiated well before the end of the 19th Century. The work continued at a slow but steady pace throughout the first half of the 20th Century. Findings from the effort had little impact on medicine and other aspects of human biology, however, until the 6th decade of the 20th Century. During that decade, research in the area was begun by eight groups of investigators, each of which was led by one or two senior scientists with great experimental talent, creativity and foresight. Their findings added new dimension to knowledge of the microbiota and initiated an explosion of interest in research in the field that has continued to the present day. The research of the groups during the 1960's is described in this review as a tribute to the senior scientists who had such critical impact on this important field of study.
- 15Pariente, N. A field is born. 2019. https://www.nature.com/articles/d42859-019-00006-2.Google ScholarThere is no corresponding record for this reference.
- 16Micheli, P. A. Nova plantarum genera iuxta Tournefortii methodum disposita ; 1729.Google ScholarThere is no corresponding record for this reference.
- 17Drews, G. The roots of microbiology and the influence of Ferdinand Cohn on microbiology of the 19th century. FEMS Microbiol. Rev. 2000, 24, 225– 249, DOI: 10.1111/j.1574-6976.2000.tb00540.xGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXjslOru7g%253D&md5=402d7cf8cd6b2bf3509d72761ad9e6a9The roots of microbiology and the influence of Ferdinand Cohn on microbiology of the 19th centuryDrews, G.FEMS Microbiology Reviews (2000), 24 (3), 225-249CODEN: FMREE4; ISSN:0168-6445. (Elsevier Science B.V.)The beginning of modern microbiol. can be traced back to the 1870s, and it was based on the development of new concepts that originated during the two preceding centuries on the role of microorganisms, new exptl. methods, and discoveries in chem., physics, and evolutionary cell biol. The crucial progress was the isolation and growth on solid media of clone cultures arising from single cells and the demonstration that these pure cultures have specific, inheritable characteristics and metabolic capacities. The doctrine of the spontaneous generation of microorganisms, which stimulated research for a century, lost its role as an important concept. Microorganisms were discovered to be causative agents of infectious diseases and of specific metabolic processes. Microscopy techniques advanced studies on microorganisms. The discovery of sexuality and development in microorganisms and Darwin's theory of evolution contributed to the founding of microbiol. as a science. Ferdinand Cohn (1828-1898), a pioneer in the developmental biol. of lower plants, considerably promoted the taxonomy and physiol. of bacteria, discovered the heat-resistant endospores of bacilli, and was active in applied microbiol.
- 18Boundless. Koch’s Postulates. https://bio.libretexts.org/Bookshelves/Microbiology/Book%3A_Microbiology_(Boundless)/10%3A_Epidemiology/10.1%3A_Principles_of_Epidemiology/10.1D%3A__Kochs_Postulates (accessed October 6, 2022).Google ScholarThere is no corresponding record for this reference.
- 19Metchnikoff, E. Prolongation of Life: Optimistic Studies ; 1923.Google ScholarThere is no corresponding record for this reference.
- 20Sonnenborn, U. Escherichia coli strain Nissle 1917─from bench to bedside and back: history of a special Escherichia coli strain with probiotic properties. FEMS Microbiol. Lett. 2016, 363, fnw212, DOI: 10.1093/femsle/fnw212Google ScholarThere is no corresponding record for this reference.
- 21Tan, S. Y.; Tatsumura, Y. Alexander Fleming (1881–1955): Discoverer of penicillin. Singapore Med. J. 2015, 56, 366– 367, DOI: 10.11622/smedj.2015105Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC287gtVKisQ%253D%253D&md5=49579a42a8642f0f8f09c8c49e00a60eAlexander Fleming (1881-1955): Discoverer of penicillinTan Siang Yong; Tatsumura YvonneSingapore medical journal (2015), 56 (7), 366-7 ISSN:0037-5675.There is no expanded citation for this reference.
- 22Almeida, A.; Mitchell, A. L.; Boland, M.; Forster, S. C.; Gloor, G. B.; Tarkowska, A.; Lawley, T. D.; Finn, R. D. A new genomic blueprint of the human gut microbiota. Nature 2019, 568, 499– 504, DOI: 10.1038/s41586-019-0965-1Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmslKhu7s%253D&md5=4871997c22d8542f9df02ad2c0066e61A new genomic blueprint of the human gut microbiotaAlmeida, Alexandre; Mitchell, Alex L.; Boland, Miguel; Forster, Samuel C.; Gloor, Gregory B.; Tarkowska, Aleksandra; Lawley, Trevor D.; Finn, Robert D.Nature (London, United Kingdom) (2019), 568 (7753), 499-504CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The compn. of the human gut microbiota is linked to health and disease, but knowledge of individual microbial species is needed to decipher their biol. roles. Despite extensive culturing and sequencing efforts, the complete bacterial repertoire of the human gut microbiota remains undefined. We identify 1952 uncultured candidate bacterial species by reconstructing 92,143 metagenome-assembled genomes from 11,850 human gut microbiomes. These uncultured genomes substantially expand the known species repertoire of the collective human gut microbiota, with a 281% increase in phylogenetic diversity. Although the newly identified species are less prevalent in well-studied populations compared to ref. isolate genomes, they improve classification of understudied African and South American samples by >200%. These candidate species encode hundreds of newly identified biosynthetic gene clusters and possess a distinctive functional capacity that might explain their elusive nature. Our work expands the known diversity of uncultured gut bacteria, which provides unprecedented resoln. for taxonomic and functional characterization of the intestinal microbiota.
- 23Hungate, R. E., Chapter IV A Roll Tube Method for Cultivation of Strict Anaerobes. In Methods in Microbiology, Vol. 3; Norris, J. R., Ribbons, D. W., Eds.; Academic Press, 1969; pp 117– 132.Google ScholarThere is no corresponding record for this reference.
- 24Cat, L. A. The Decade Of The Microbiome. https://www.forbes.com/sites/linhanhcat/2019/12/31/decade-of-the-microbiome/?sh=7e621a679961 (accessed October 11, 2022).Google ScholarThere is no corresponding record for this reference.
- 25NIH Human Microbiome Project. https://www.hmpdacc.org/overview/ (accessed October 6, 2022).Google ScholarThere is no corresponding record for this reference.
- 26MetaHIT. https://www.gutmicrobiotaforhealth.com/metahit/ (accessed October 6, 2022).Google ScholarThere is no corresponding record for this reference.
- 27Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K. S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59– 65, DOI: 10.1038/nature08821Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXislahsLc%253D&md5=7116690033a12ae47bbe53c77f768e91A human gut microbial gene catalogue established by metagenomic sequencingQin, Junjie; Li, Ruiqiang; Raes, Jeroen; Arumugam, Manimozhiyan; Burgdorf, Kristoffer Solvsten; Manichanh, Chaysavanh; Nielsen, Trine; Pons, Nicolas; Levenez, Florence; Yamada, Takuji; Mende, Daniel R.; Li, Junhua; Xu, Junming; Li, Shaochuan; Li, Dongfang; Cao, Jianjun; Wang, Bo; Liang, Huiqing; Zheng, Huisong; Xie, Yinlong; Tap, Julien; Lepage, Patricia; Bertalan, Marcelo; Batto, Jean-Michel; Hansen, Torben; Le Paslier, Denis; Linneberg, Allan; Nielsen, H. Bjoern; Pelletier, Eric; Renault, Pierre; Sicheritz-Ponten, Thomas; Turner, Keith; Zhu, Hongmei; Yu, Chang; Li, Shengting; Jian, Min; Zhou, Yan; Li, Yingrui; Zhang, Xiuqing; Li, Songgang; Qin, Nan; Yang, Huanming; Wang, Jian; Brunak, Soeren; Dore, Joel; Guarner, Francisco; Kristiansen, Karsten; Pedersen, Oluf; Parkhill, Julian; Weissenbach, Jean; Antolin, Maria; Artiguenave, Francois; Blottiere, Herve; Borruel, Natalia; Bruls, Thomas; Casellas, Francesc; Chervaux, Christian; Cultrone, Antonella; Delorme, Christine; Denariaz, Gerard; Dervyn, Rozenn; Forte, Miguel; Friss, Carsten; van de Guchte, Maarten; Guedon, Eric; Haimet, Florence; Jamet, Alexandre; Juste, Catherine; Kaci, Ghalia; Kleerebezem, Michiel; Knol, Jan; Kristensen, Michel; Layec, Severine; Le Roux, Karine; Leclerc, Marion; Maguin, Emmanuelle; Melo Minardi, Raquel; Oozeer, Raish; Rescigno, Maria; Sanchez, Nicolas; Tims, Sebastian; Torrejon, Toni; Varela, Encarna; de Vos, Willem; Winogradsky, Yohanan; Zoetendal, Erwin; Bork, Peer; Ehrlich, S. Dusko; Wang, JunNature (London, United Kingdom) (2010), 464 (7285), 59-65CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)To understand the impact of gut microbes on human health and well-being it is crucial to assess their genetic potential. This report describes the Illumina-based metagenomic sequencing, assembly, and characterization of 3.3 million non-redundant microbial genes, derived from 576.7 gigabases of sequence, from fecal samples of 124 European individuals. The gene set, ∼150 times larger than the human gene complement, contains an overwhelming majority of the prevalent (more frequent) microbial genes of the cohort and probably includes a large proportion of the prevalent human intestinal microbial genes. The genes are largely shared among individuals of the cohort. Over 99% of the genes are bacterial, indicating that the entire cohort harbors between 1000 and 1150 prevalent bacterial species and each individual at least 160 such species, which are also largely shared. The minimal gut metagenome and the minimal gut bacterial genome are defined and described in terms of functions present in all individuals and most bacteria, resp. Sequence data are deposited in the European Short Read Archive with accession no. ERA000116.
- 28Proctor, Lita M. The Human Microbiome Project in 2011 and Beyond. Cell Host Microbe 2011, 10, 287– 291, DOI: 10.1016/j.chom.2011.10.001Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlKisbbL&md5=ca57d3202f3891c261a4b3c7b7d449daThe Human Microbiome Project in 2011 and BeyondProctor, Lita M.Cell Host & Microbe (2011), 10 (4), 287-291CODEN: CHMECB; ISSN:1931-3128. (Cell Press)A review. The human microbiome comprises the genes and genomes of the microbiota that inhabit the body. The authors highlight Human Microbiome Project (HMP) resources, including 600 microbial ref. genomes, 70 million 16S sequences, 700 metagenomes, and 60 million predicted genes from healthy adult microbiomes. Microbiome studies of specific diseases and future research directions are also discussed.
- 29Birney, E.; Stamatoyannopoulos, J. A.; Dutta, A.; Guigó, R.; Gingeras, T. R.; Margulies, E. H.; Weng, Z.; Snyder, M.; Dermitzakis, E. T.; Stamatoyannopoulos, J. A. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007, 447, 799– 816, DOI: 10.1038/nature05874Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXms1Wjsb0%253D&md5=727de4c5a83d3f75af54e31f974472a7Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot projectBirney, Ewan; Stamatoyannopoulos, John A.; Dutta, Anindya; Guigo, Roderic; Gingeras, Thomas R.; Margulies, Elliott H.; Weng, Zhiping; Snyder, Michael; Dermitzakis, Emmanouil T.; Stamatoyannopoulos, John A.; Thurman, Robert E.; Kuehn, Michael S.; Taylor, Christopher M.; Neph, Shane; Koch, Christoph M.; Asthana, Saurabh; Malhotra, Ankit; Adzhubei, Ivan; Greenbaum, Jason A.; Andrews, Robert M.; Flicek, Paul; Boyle, Patrick J.; Cao, Hua; Carter, Nigel P.; Clelland, Gayle K.; Davis, Sean; Day, Nathan; Dhami, Pawandeep; Dillon, Shane C.; Dorschner, Michael O.; Fiegler, Heike; Giresi, Paul G.; Goldy, Jeff; Hawrylycz, Michael; Haydock, Andrew; Humbert, Richard; James, Keith D.; Johnson, Brett E.; Johnson, Ericka M.; Frum, Tristan T.; Rosenzweig, Elizabeth R.; Karnani, Neerja; Lee, Kirsten; Lefebvre, Gregory C.; Navas, Patrick A.; Neri, Fidencio; Parker, Stephen C. J.; Sabo, Peter J.; Sandstrom, Richard; Shafer, Anthony; Vetrie, David; Weaver, Molly; Wilcox, Sarah; Yu, Man; Collins, Francis S.; Dekker, Job; Lieb, Jason D.; Tullius, Thomas D.; Crawford, Gregory E.; Sunyaev, Shamil; Noble, William S.; Dunham, Ian; Dutta, Anindya; Guigo, Roderic; Denoeud, France; Reymond, Alexandre; Kapranov, Philipp; Rozowsky, Joel; Zheng, Deyou; Castelo, Robert; Frankish, Adam; Harrow, Jennifer; Ghosh, Srinka; Sandelin, Albin; Hofacker, Ivo L.; Baertsch, Robert; Keefe, Damian; Flicek, Paul; Dike, Sujit; Cheng, Jill; Hirsch, Heather A.; Sekinger, Edward A.; Lagarde, Julien; Abril, Josep F.; Shahab, Atif; Flamm, Christoph; Fried, Claudia; Hackermueller, Joerg; Hertel, Jana; Lindemeyer, Manja; Missal, Kristin; Tanzer, Andrea; Washietl, Stefan; Korbel, Jan; Emanuelsson, Olof; Pedersen, Jakob S.; Holroyd, Nancy; Taylor, Ruth; Swarbreck, David; Matthews, Nicholas; Dickson, Mark C.; Thomas, Daryl J.; Weirauch, Matthew T.; Gilbert, James; Drenkow, Jorg; Bell, Ian; Zhao, Xiao Dong; Srinivasan, K. G.; Sung, Wing-Kin; Ooi, Hong Sain; Chiu, Kuo Ping; Foissac, Sylvain; Alioto, Tyler; Brent, Michael; Pachter, Lior; Tress, Michael L.; Valencia, Alfonso; Choo, Siew Woh; Choo, Chiou Yu; Ucla, Catherine; Manzano, Caroline; Wyss, Carine; Cheung, Evelyn; Clark, Taane G.; Brown, James B.; Ganesh, Madhavan; Patel, Sandeep; Tammana, Hari; Chrast, Jacqueline; Henrichsen, Charlotte N.; Kai, Chikatoshi; Kawai, Jun; Nagalakshmi, Ugrappa; Wu, Jiaqian; Lian, Zheng; Lian, Jin; Newburger, Peter; Zhang, Xueqing; Bickel, Peter; Mattick, John S.; Carninci, Piero; Hayashizaki, Yoshihide; Weissman, Sherman; Dermitzakis, Emmanouil T.; Margulies, Elliott H.; Hubbard, Tim; Myers, Richard M.; Rogers, Jane; Stadler, Peter F.; Lowe, Todd M.; Wei, Chia-Lin; Ruan, Yijun; Snyder, Michael; Birney, Ewan; Struhl, Kevin; Gerstein, Mark; Antonarakis, Stylianos E.; Gingeras, Thomas R.; Brown, James B.; Flicek, Paul; Fu, Yutao; Keefe, Damian; Birney, Ewan; Denoeud, France; Gerstein, Mark; Green, Eric D.; Kapranov, Philipp; Karaoez, Ulas; Myers, Richard M.; Noble, William S.; Reymond, Alexandre; Rozowsky, Joel; Struhl, Kevin; Siepel, Adam; Stamatoyannopoulos, John A.; Taylor, Christopher M.; Taylor, James; Thurman, Robert E.; Tullius, Thomas D.; Washietl, Stefan; Zheng, Deyou; Liefer, Laura A.; Wetterstrand, Kris A.; Good, Peter J.; Feingold, Elise A.; Guyer, Mark S.; Collins, Francis S.; Margulies, Elliott H.; Cooper, Gregory M.; Asimenos, George; Thomas, Daryl J.; Dewey, Colin N.; Siepel, Adam; Birney, Ewan; Keefe, Damian; Hou, Minmei; Taylor, James; Nikolaev, Sergey; Montoya-Burgos, Juan I.; Loeytynoja, Ari; Whelan, Simon; Pardi, Fabio; Massingham, Tim; Brown, James B.; Huang, Haiyan; Zhang, Nancy R.; Bickel, Peter; Holmes, Ian; Mullikin, James C.; Ureta-Vidal, Abel; Paten, Benedict; Seringhaus, Michael; Church, Deanna; Rosenbloom, Kate; Kent, W. James; Stone, Eric A.; Gerstein, Mark; Antonarakis, Stylianos E.; Batzoglou, Serafim; Goldman, Nick; Hardison, Ross C.; Haussler, David; Miller, Webb; Pachter, Lior; Green, Eric D.; Sidow, Arend; Weng, Zhiping; Trinklein, Nathan D.; Fu, Yutao; Zhang, Zhengdong D.; Karaoez, Ulas; Barrera, Leah; Stuart, Rhona; Zheng, Deyou; Ghosh, Srinka; Flicek, Paul; King, David C.; Taylor, James; Ameur, Adam; Enroth, Stefan; Bieda, Mark C.; Koch, Christoph M.; Hirsch, Heather A.; Wei, Chia-Lin; Cheng, Jill; Kim, Jonghwan; Bhinge, Akshay A.; Giresi, Paul G.; Jiang, Nan; Liu, Jun; Yao, Fei; Sung, Wing-Kin; Chiu, Kuo Ping; Vega, Vinsensius B.; Lee, Charlie W. H.; Ng, Patrick; Shahab, Atif; Sekinger, Edward A.; Yang, Annie; Moqtaderi, Zarmik; Zhu, Zhou; Xu, Xiaoqin; Squazzo, Sharon; Oberley, Matthew J.; Inman, David; Singer, Michael A.; Richmond, Todd A.; Munn, Kyle J.; Rada-Iglesias, Alvaro; Wallerman, Ola; Komorowski, Jan; Clelland, Gayle K.; Wilcox, Sarah; Dillon, Shane C.; Andrews, Robert M.; Fowler, Joanna C.; Couttet, Phillippe; James, Keith D.; Lefebvre, Gregory C.; Bruce, Alexander W.; Dovey, Oliver M.; Ellis, Peter D.; Dhami, Pawandeep; Langford, Cordelia F.; Carter, Nigel P.; Vetrie, David; Kapranov, Philipp; Nix, David A.; Bell, Ian; Patel, Sandeep; Rozowsky, Joel; et al.Nature (London, United Kingdom) (2007), 447 (7146), 799-816CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)We report the generation and anal. of functional data from multiple, diverse expts. performed on a targeted 1% of the human genome as part of the pilot phase of the ENCODE Project. These data have been further integrated and augmented by a no. of evolutionary and computational analyses. Together, our results advance the collective knowledge about human genome function in several major areas. First, our studies provide convincing evidence that the genome is pervasively transcribed, such that the majority of its bases can be found in primary transcripts, including non-protein-coding transcripts, and those that extensively overlap one another. Second, systematic examn. of transcriptional regulation has yielded new understanding about transcription start sites, including their relationship to specific regulatory sequences and features of chromatin accessibility and histone modification. Third, a more sophisticated view of chromatin structure has emerged, including its inter-relationship with DNA replication and transcriptional regulation. Finally, integration of these new sources of information, in particular with respect to mammalian evolution based on inter- and intra-species sequence comparisons, has yielded new mechanistic and evolutionary insights concerning the functional landscape of the human genome. Together, these studies are defining a path for pursuit of a more comprehensive characterization of human genome function.
- 30Riesenfeld, C. S.; Schloss, P. D.; Handelsman, J. Metagenomics: Genomic Analysis of Microbial Communities. Annu. Rev. Genet. 2004, 38, 525– 552, DOI: 10.1146/annurev.genet.38.072902.091216Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXltlyjtg%253D%253D&md5=84d6562bb905baacfe91982a2151722dMetagenomics: Genomic analysis of microbial communitiesRiesenfeld, Christian S.; Schloss, Patrick D.; Handelsman, JoAnnual Review of Genetics (2004), 38 (), 525-552CODEN: ARVGB7; ISSN:0066-4197. (Annual Reviews Inc.)A review. Uncultured microorganisms comprise the majority of the planet's biol. diversity. Microorganisms represent two of the three domains of life and contain vast diversity that is the product of an estd. 3.8 billion years of evolution. In many environments, as many as 99% of the microorganisms cannot be cultured by std. techniques, and the uncultured fraction includes diverse organisms that are only distantly related to the cultured ones. Therefore, culture-independent methods are essential to understand the genetic diversity, population structure, and ecol. roles of the majority of microorganisms. Metagenomics, or the culture-independent genomic anal. of an assemblage of microorganisms, has potential to answer fundamental questions in microbial ecol. This review describes progress toward understanding the biol. of uncultured Bacteria, Archaea, and viruses through metagenomic analyses.
- 31Jandhyala, S. M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787– 8803, DOI: 10.3748/wjg.v21.i29.8787Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XosVSlsw%253D%253D&md5=b0cd06c0ec390547e6248afb8e782139Role of the normal gut microbiotaJandhyala, Sai Manasa; Talukdar, Rupjyoti; Subramanyam, Chivkula; Vuyyuru, Harish; Sasikala, Mitnala; Reddy, D. NageshwarWorld Journal of Gastroenterology (2015), 21 (29), 8787-8803CODEN: WJGAF2; ISSN:2219-2840. (Baishideng Publishing Group Inc.)Relation between the gut microbiota and human health is being increasingly recognized. It is now well established that a healthy gut flora is largely responsible for overall health of the host. The normal human gut microbiota comprises of two major phyla, namely Bacteroidetes and Firmicutes. Though the gut microbiota in an infant appears haphazard, it starts resembling the adult flora by the age of 3 years. Nevertheless, there exist temporal and spatial variations in the microbial distribution from esophagus to the rectum all along the individual's life span. Developments in genome sequencing technologies and bioinformatics have now enabled scientists to study these microorganisms and their function and microbehost interactions in an elaborate manner both in health and disease. The normal gut microbiota imparts specific function in host nutrient metab., xenobiotic and drug metab., maintenance of structural integrity of the gut mucosal barrier, immunomodulation, and protection against pathogens. Several factors play a role in shaping the normal gut microbiota. They include (1) the mode of delivery (vaginal or caesarean); (2) diet during infancy (breast milk or formula feeds) and adulthood (vegan based or meat based); and (3) use of antibiotics or antibiotic like mols. that are derived from the environment or the gut commensal community. A major concern of antibiotic use is the long-term alteration of the normal healthy gut microbiota and horizontal transfer of resistance genes that could result in reservoir of organisms with a multidrug resistant gene pool.
- 32Tomas, J.; Wrzosek, L.; Bouznad, N.; Bouet, S.; Mayeur, C.; Noordine, M. L.; Honvo-Houeto, E.; Langella, P.; Thomas, M.; Cherbuy, C. Primocolonization is associated with colonic epithelial maturation during conventionalization. FASEB J. 2013, 27, 645– 655, DOI: 10.1096/fj.12-216861Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXit1Skur0%253D&md5=311f2c67cbf2a56fdc02893385a4190bPrimocolonization is associated with colonic epithelial maturation during conventionalizationTomas, Julie; Wrzosek, Laura; Bouznad, Nassim; Bouet, Stephan; Mayeur, Camille; Noordine, Marie-Louise; Honvo-Houeto, Edith; Langella, Philippe; Thomas, Muriel; Cherbuy, ClaireFASEB Journal (2013), 27 (2), 645-655, 10.1096/fj.12-216861CODEN: FAJOEC; ISSN:0892-6638. (Federation of American Societies for Experimental Biology)Interaction between the gut microbiota and the host starts immediately after birth with the progressive colonization of the sterile intestine. Our aim was to investigate the interactions taking place in the colonic epithelium after the first exposure to gut microbiota. Germ-free (GF) rats were inoculated with two different microbiotas: the first, obtained from suckling rats, was rich in primocolonizing bacteria and the second, obtained from adult rats, was representative of a mature microbiota. Once transferred into GF rats, these two microbiotas evolved such that they converged, and recapitulated the primocolonization pattern, mimicking the chronol. scheme of implantation following birth. The two microbiotas induced common responses in the colonic epithelium: a transitory proliferative phase followed by a compensatory phase characterized by increases in the abundance of p21Cip1 and p27Kip1 and in the no. of goblet cells. The effects of the two microbiotas diverged only through their effects on colonic transporters. Analyses of solute carriers and aquaporins revealed that functional maturation was more pronounced following exposure to adult microbiota than suckling microbiota. The colon matured in parallel with the evolution of the microbiota compn., and we therefore suggest a link between intestinal events regulating homeostasis of the colon and modulation of microbial compn.
- 33Caballero, S.; Pamer, E. G. Microbiota-mediated inflammation and antimicrobial defense in the intestine. Annu. Rev. Immunol. 2015, 33, 227– 256, DOI: 10.1146/annurev-immunol-032713-120238Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVSisL3M&md5=9f613a189ee2cb8447e3e7330eb4bc4dMicrobiota-Mediated Inflammation and Antimicrobial Defense in the IntestineCaballero, Silvia; Pamer, Eric G.Annual Review of Immunology (2015), 33 (), 227-256CODEN: ARIMDU; ISSN:0732-0582. (Annual Reviews)The diverse microbial populations constituting the intestinal microbiota promote immune development and differentiation, but because of their complex metabolic requirements and the consequent difficulty culturing them, they remained, until recently, largely uncharacterized and mysterious. In the last decade, deep nucleic acid sequencing platforms, new computational and bioinformatics tools, and full-genome characterization of several hundred commensal bacterial species facilitated studies of the microbiota and revealed that differences in microbiota compn. can be assocd. with inflammatory, metabolic, and infectious diseases, that each human is colonized by a distinct bacterial flora, and that the microbiota can be manipulated to reduce and even cure some diseases. Different bacterial species induce distinct immune cell populations that can play pro- and anti-inflammatory roles, and thus the compn. of the microbiota dets., in part, the level of resistance to infection and susceptibility to inflammatory diseases. This review summarizes recent work characterizing commensal microbes that contribute to the antimicrobial defense/inflammation axis.
- 34Magnúsdóttir, S.; Ravcheev, D.; de Crécy-Lagard, V.; Thiele, I. Systematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbes. Frontiers in Genetics 2015, 6, 148, DOI: 10.3389/fgene.2015.00148Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MfgvVWltA%253D%253D&md5=4f500bfea9a3f72d0e88539d7e3ccfcfSystematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbesMagnusdottir Stefania; Ravcheev Dmitry; Thiele Ines; de Crecy-Lagard ValerieFrontiers in genetics (2015), 6 (), 148 ISSN:1664-8021.The human gut microbiota supplies its host with essential nutrients, including B-vitamins. Using the PubSEED platform, we systematically assessed the genomes of 256 common human gut bacteria for the presence of biosynthesis pathways for eight B-vitamins: biotin, cobalamin, folate, niacin, pantothenate, pyridoxine, riboflavin, and thiamin. On the basis of the presence and absence of genome annotations, we predicted that each of the eight vitamins was produced by 40-65% of the 256 human gut microbes. The distribution of synthesis pathways was diverse; some genomes had all eight biosynthesis pathways, whereas others contained no de novo synthesis pathways. We compared our predictions to experimental data from 16 organisms and found 88% of our predictions to be in agreement with published data. In addition, we identified several pairs of organisms whose vitamin synthesis pathway pattern complemented those of other organisms. This analysis suggests that human gut bacteria actively exchange B-vitamins among each other, thereby enabling the survival of organisms that do not synthesize any of these essential cofactors. This result indicates the co-evolution of the gut microbes in the human gut environment. Our work presents the first comprehensive assessment of the B-vitamin synthesis capabilities of the human gut microbiota. We propose that in addition to diet, the gut microbiota is an important source of B-vitamins, and that changes in the gut microbiota composition can severely affect our dietary B-vitamin requirements.
- 35Mayer, E. A.; Knight, R.; Mazmanian, S. K.; Cryan, J. F.; Tillisch, K. Gut microbes and the brain: paradigm shift in neuroscience. J. Neurosci. 2014, 34, 15490– 15496, DOI: 10.1523/JNEUROSCI.3299-14.2014Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2M3nvVKktw%253D%253D&md5=94069bb4e4d54c2afed184e28851a2c6Gut microbes and the brain: paradigm shift in neuroscienceMayer Emeran A; Knight Rob; Mazmanian Sarkis K; Cryan John F; Tillisch KirstenThe Journal of neuroscience : the official journal of the Society for Neuroscience (2014), 34 (46), 15490-6 ISSN:.The discovery of the size and complexity of the human microbiome has resulted in an ongoing reevaluation of many concepts of health and disease, including diseases affecting the CNS. A growing body of preclinical literature has demonstrated bidirectional signaling between the brain and the gut microbiome, involving multiple neurocrine and endocrine signaling mechanisms. While psychological and physical stressors can affect the composition and metabolic activity of the gut microbiota, experimental changes to the gut microbiome can affect emotional behavior and related brain systems. These findings have resulted in speculation that alterations in the gut microbiome may play a pathophysiological role in human brain diseases, including autism spectrum disorder, anxiety, depression, and chronic pain. Ongoing large-scale population-based studies of the gut microbiome and brain imaging studies looking at the effect of gut microbiome modulation on brain responses to emotion-related stimuli are seeking to validate these speculations. This article is a summary of emerging topics covered in a symposium and is not meant to be a comprehensive review of the subject.
- 36Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R. D.; Shanahan, F.; Dinan, T. G.; Cryan, J. F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666– 673, DOI: 10.1038/mp.2012.77Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXot1aku7w%253D&md5=46c3ff1746ad758d4af322c058f8b4cbThe microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent mannerClarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R. D.; Shanahan, F.; Dinan, T. G.; Cryan, J. F.Molecular Psychiatry (2013), 18 (6), 666-673CODEN: MOPSFQ; ISSN:1359-4184. (Nature Publishing Group)Bacterial colonization of the intestine has a major role in the post-natal development and maturation of the immune and endocrine systems. These processes are key factors underpinning central nervous system (CNS) signalling. Regulation of the microbiome-gut-brain axis is essential for maintaining homeostasis, including that of the CNS. However, there is a paucity of data pertaining to the influence of microbiome on the serotonergic system. Germ-free (GF) animals represent an effective preclin. tool to investigate such phenomena. Here we show that male GF animals have a significant elevation in the hippocampal concn. of 5-hydroxytryptamine and 5-hydroxyindoleacetic acid, its main metabolite, compared with conventionally colonized control animals. Moreover, this alteration is sex specific in contrast with the immunol. and neuroendocrine effects which are evident in both sexes. Concns. of tryptophan, the precursor of serotonin, are increased in the plasma of male GF animals, suggesting a humoral route through which the microbiota can influence CNS serotonergic neurotransmission. Interestingly, colonization of the GF animals post weaning is insufficient to reverse the CNS neurochem. consequences in adulthood of an absent microbiota in early life despite the peripheral availability of tryptophan being restored to baseline values. In addn., reduced anxiety in GF animals is also normalized following restoration of the intestinal microbiota. These results demonstrate that CNS neurotransmission can be profoundly disturbed by the absence of a normal gut microbiota and that this aberrant neurochem., but not behavioral, profile is resistant to restoration of a normal gut flora in later life. Mol. Psychiatry (2013) 18, 666-673; doi:10.1038/mp.2012.77; published online 12 June 2012.
- 37Bray, N. The microbiota–gut–brain axis. https://www.nature.com/articles/d42859-019-00021-3 (accessed October 6, 2022).Google ScholarThere is no corresponding record for this reference.
- 38Dinan, T. G.; Cryan, J. F. The impact of gut microbiota on brain and behaviour: implications for psychiatry. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 552– 558, DOI: 10.1097/MCO.0000000000000221Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC283itlCitw%253D%253D&md5=66e25889e8960eb947e2384e8782ebb3The impact of gut microbiota on brain and behaviour: implications for psychiatryDinan Timothy G; Cryan John FCurrent opinion in clinical nutrition and metabolic care (2015), 18 (6), 552-8 ISSN:.PURPOSE OF REVIEW: The gut microbiota has become a focus of research for those interested in the brain and behaviour. Here, we profile the gut microbiota in a variety of neuropsychiatric syndromes. RECENT FINDINGS: Multiple routes of communication between the gut and brain have been established and these include the vagus nerve, immune system, short chain fatty acids and tryptophan. Developmentally, those born by caesarean section have a distinctly different microbiota in early life to those born per vaginum. At the other extreme, individuals who age with considerable ill-heath tend to show narrowing in microbial diversity. Recently, the gut microbiota has been profiled in a variety of conditions including autism, major depression and Parkinson's disease. There is still debate as to whether or not these changes are core to the pathophysiology or merely epiphenomenal. SUMMARY: The current narrative suggests that certain neuropsychiatric disorders might be treated by targeting the microbiota either by microbiota transplantation, antibiotics or psychobiotics.
- 39Bravo, J. A.; Forsythe, P.; Chew, M. V.; Escaravage, E.; Savignac, H. M.; Dinan, T. G.; Bienenstock, J.; Cryan, J. F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 16050– 16055, DOI: 10.1073/pnas.1102999108Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1ekt7nF&md5=f87375fac8abc7761ef696afb9b42a59Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerveBravo, Javier A.; Forsythe, Paul; Chew, Marianne V.; Escaravage, Emily; Savignac, Helene M.; Dinan, Timothy G.; Bienenstock, John; Cryan, Ohn F.Proceedings of the National Academy of Sciences of the United States of America (2011), 108 (38), 16050-16055, S16050/1-S16050/7CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)There is increasing, but largely indirect, evidence pointing to an effect of commensal gut microbiota on the central nervous system (CNS). However, it is unknown whether lactic acid bacteria such as Lactobacillus rhamnosus could have a direct effect on neurotransmitter receptors in the CNS in normal, healthy animals. GABA is the main CNS inhibitory neurotransmitter and is significantly involved in regulating many physiol. and psychol. processes. Alterations in central GABA receptor expression are implicated in the pathogenesis of anxiety and depression, which are highly comorbid with functional bowel disorders. In this work, we show that chronic treatment with L. rhamnosus (JB-1) induced region-dependent alterations in GABAB1b mRNA in the brain with increases in cortical regions (cingulate and prelimbic) and concomitant redns. in expression in the hippocampus, amygdala, and locus coeruleus, in comparison with control-fed mice. In addn., L. rhamnosus (JB-1) reduced GABAAα2 mRNA expression in the prefrontal cortex and amygdala, but increased GABAAα2 in the hippocampus. Importantly, L. rhamnosus (JB-1) reduced stress-induced corticosterone. and anxiety- and depression-related behavior. Moreover, the neurochem. and behavioral effects were not found in vagotomized mice, identifying the vagus as a major modulatory constitutive communication pathway between the bacteria exposed to the gut and the brain. Together, these findings highlight the important role of bacteria in the bidirectional communication of the gut-brain axis and suggest that certain organisms may prove to be useful therapeutic adjuncts in stress- related disorders such as anxiety and depression.
- 40Cohen-Sacks, H.; Elazar, V.; Gao, J.; Golomb, A.; Adwan, H.; Korchov, N.; Levy, R. J.; Berger, M. R.; Golomb, G. Delivery and expression of pDNA embedded in collagen matrices. J. Controlled Release 2004, 95, 309– 320, DOI: 10.1016/j.jconrel.2003.11.001Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsVCms7w%253D&md5=7cee5279df6c140cab54b64d33b4bb13Delivery and expression of pDNA embedded in collagen matricesCohen-Sacks, Hagit; Elazar, Victoria; Gao, Jianchuan; Golomb, Assaf; Adwan, Hassan; Korchov, Nikoly; Levy, Robert J.; Berger, Martin R.; Golomb, GershonJournal of Controlled Release (2004), 95 (2), 309-320CODEN: JCREEC; ISSN:0168-3659. (Elsevier)Collagen matrixes can be used as non-viral biocompatible gene carriers for localized implantable gene therapy. Collagen matrixes embedding pDNA with enhanced binding through condensing agent linkage to the matrix or to the pDNA have been formulated, and characterized in various systems. PDNA and condensed pDNA were released intact from the matrixes within 1-2 days. In vitro transfection with collagen matrixes contg. pDNA (luciferase encoding), pDNA in liposome (LIP), and pDNA with polyethylenimine (PEI) resulted in significantly higher expression levels in comparison to naked pDNA. PDNA-LIP matrixes exhibited a dose response transfection of NIH 3T3, 293, MDA-MB-231 and smooth muscle cells (SMCs) in cell cultures. Subdermal implantations of collagen-polylysine-pDNA matrixes in rats resulted in significantly higher gene expression levels in comparison to non-condensed pDNA matrixes. Perivascular treatment with pDNA matrix and of naked pDNA soln. in balloon-injured rat carotid arteries resulted in significant expression. In conclusion, a facile method for embedding cationic formulations of pDNA in collagen matrixes was developed. These bioactive matrixes seem to be suitable for tissue engineering and local gene therapy strategies.
- 41Rutsch, A.; Kantsjö, J. B.; Ronchi, F. The Gut-Brain Axis: How Microbiota and Host Inflammasome Influence Brain Physiology and Pathology. Front. Immunol. 2020, 11, 604179, DOI: 10.3389/fimmu.2020.604179Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitVCnsL8%253D&md5=ae2dc7a2f0e92a3158a44011755e4cdcThe gut-brain axis: how microbiota and host inflammasome influence brain physiology and pathologyRutsch, Andrina; Kantsjo, Johan B.; Ronchi, FrancescaFrontiers in Immunology (2020), 11 (), 604179CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)A review. The human microbiota has a fundamental role in host physiol. and pathol. Gut microbial alteration, also known as dysbiosis, is a condition assocd. not only with gastrointestinal disorders but also with diseases affecting other distal organs. Recently it became evident that the intestinal bacteria can affect the central nervous system (CNS) physiol. and inflammation. The nervous system and the gastrointestinal tract are communicating through a bidirectional network of signaling pathways called the gutbrain axis, which consists of multiple connections, including the vagus nerve, the immune system, and bacterial metabolites and products. During dysbiosis, these pathways are dysregulated and assocd. with altered permeability of the blood-brain barrier (BBB) and neuroinflammation. However, numerous mechanisms behind the impact of the gut microbiota in neuro-development and -pathogenesis remain poorly understood. There are several immune pathways involved in CNS homeostasis and inflammation. Among those, the inflammasome pathway has been linked to neuroinflammatory conditions such as multiple sclerosis, Alzheimer's and Parkinson's diseases, but also anxiety and depressivelike disorders. The inflammasome complex assembles upon cell activation due to exposure to microbes, danger signals, or stress and lead to the prodn. of proinflammatory cytokines (interleukin-1b and interleukin-1β) and to pyroptosis. Evidences suggest that there is a reciprocal influence of microbiota and inflammasome activation in the brain. However, how this influence is precisely working is yet to be discovered. Herein, we discuss the status of the knowledge and the open questions in the field focusing on the function of intestinal microbial metabolites or products on CNS cells during healthy and inflammatory conditions, such as multiple sclerosis, Alzheimer's and Parkinson's diseases, and also neuropsychiatric disorders. In particular, we focus on the innate inflammasome pathway as immune mechanism that can be involved in several of these conditions, upon exposure to certain microbes.
- 42Drossman, D. A.; Hasler, W. L. Rome IV-Functional GI Disorders: Disorders of Gut-Brain Interaction. Gastroenterology 2016, 150, 1257– 1261, DOI: 10.1053/j.gastro.2016.03.035Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28bmsFyhsQ%253D%253D&md5=27727046d3fb8623210eb13dcae74c85Rome IV-Functional GI Disorders: Disorders of Gut-Brain InteractionDrossman Douglas A; Hasler William LGastroenterology (2016), 150 (6), 1257-61 ISSN:.There is no expanded citation for this reference.
- 43Perez-Muñoz, M. E.; Arrieta, M.-C.; Ramer-Tait, A. E.; Walter, J. A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: implications for research on the pioneer infant microbiome. Microbiome 2017, 5, 48, DOI: 10.1186/s40168-017-0268-4Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1critlWquw%253D%253D&md5=ed193fdc5a2e0d6896d48b5d74a43cd7A critical assessment of the "sterile womb" and "in utero colonization" hypotheses: implications for research on the pioneer infant microbiomePerez-Munoz Maria Elisa; Walter Jens; Arrieta Marie-Claire; Arrieta Marie-Claire; Ramer-Tait Amanda E; Walter JensMicrobiome (2017), 5 (1), 48 ISSN:.After more than a century of active research, the notion that the human fetal environment is sterile and that the neonate's microbiome is acquired during and after birth was an accepted dogma. However, recent studies using molecular techniques suggest bacterial communities in the placenta, amniotic fluid, and meconium from healthy pregnancies. These findings have led many scientists to challenge the "sterile womb paradigm" and propose that microbiome acquisition instead begins in utero, an idea that would fundamentally change our understanding of gut microbiota acquisition and its role in human development. In this review, we provide a critical assessment of the evidence supporting these two opposing hypotheses, specifically as it relates to (i) anatomical, immunological, and physiological characteristics of the placenta and fetus; (ii) the research methods currently used to study microbial populations in the intrauterine environment; (iii) the fecal microbiome during the first days of life; and (iv) the generation of axenic animals and humans. Based on this analysis, we argue that the evidence in support of the "in utero colonization hypothesis" is extremely weak as it is founded almost entirely on studies that (i) used molecular approaches with an insufficient detection limit to study "low-biomass" microbial populations, (ii) lacked appropriate controls for contamination, and (iii) failed to provide evidence of bacterial viability. Most importantly, the ability to reliably derive axenic animals via cesarean sections strongly supports sterility of the fetal environment in mammals. We conclude that current scientific evidence does not support the existence of microbiomes within the healthy fetal milieu, which has implications for the development of clinical practices that prevent microbiome perturbations after birth and the establishment of future research priorities.
- 44Dominguez-Bello, M. G.; Costello, E. K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. U. S. A. 2010, 107, 11971– 11975, DOI: 10.1073/pnas.1002601107Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3cnlslKksg%253D%253D&md5=0aaa3ce83c3d727862accea6cbc2401aDelivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newbornsDominguez-Bello Maria G; Costello Elizabeth K; Contreras Monica; Magris Magda; Hidalgo Glida; Fierer Noah; Knight RobProceedings of the National Academy of Sciences of the United States of America (2010), 107 (26), 11971-5 ISSN:.Upon delivery, the neonate is exposed for the first time to a wide array of microbes from a variety of sources, including maternal bacteria. Although prior studies have suggested that delivery mode shapes the microbiota's establishment and, subsequently, its role in child health, most researchers have focused on specific bacterial taxa or on a single body habitat, the gut. Thus, the initiation stage of human microbiome development remains obscure. The goal of the present study was to obtain a community-wide perspective on the influence of delivery mode and body habitat on the neonate's first microbiota. We used multiplexed 16S rRNA gene pyrosequencing to characterize bacterial communities from mothers and their newborn babies, four born vaginally and six born via Cesarean section. Mothers' skin, oral mucosa, and vagina were sampled 1 h before delivery, and neonates' skin, oral mucosa, and nasopharyngeal aspirate were sampled <5 min, and meconium <24 h, after delivery. We found that in direct contrast to the highly differentiated communities of their mothers, neonates harbored bacterial communities that were undifferentiated across multiple body habitats, regardless of delivery mode. Our results also show that vaginally delivered infants acquired bacterial communities resembling their own mother's vaginal microbiota, dominated by Lactobacillus, Prevotella, or Sneathia spp., and C-section infants harbored bacterial communities similar to those found on the skin surface, dominated by Staphylococcus, Corynebacterium, and Propionibacterium spp. These findings establish an important baseline for studies tracking the human microbiome's successional development in different body habitats following different delivery modes, and their associated effects on infant health.
- 45Hill, C. J.; Lynch, D. B.; Murphy, K.; Ulaszewska, M.; Jeffery, I. B.; O’Shea, C. A.; Watkins, C.; Dempsey, E.; Mattivi, F.; Tuohy, K. Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET Cohort. Microbiome 2017, 5, 4, DOI: 10.1186/s40168-016-0213-yGoogle Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1c7msFykug%253D%253D&md5=37ab7baa46268117464df0482f93e01aEvolution of gut microbiota composition from birth to 24 weeks in the INFANTMET CohortHill Cian J; Lynch Denise B; Murphy Kiera; Jeffery Ian B; Ross R Paul; O' Toole Paul W; Hill Cian J; Lynch Denise B; Murphy Kiera; Ross R Paul; Ryan C Anthony; O' Toole Paul W; Stanton Catherine; Murphy Kiera; Watkins Claire; Stanton Catherine; Ulaszewska Marynka; Mattivi Fulvio; Tuohy Kieran; O'Shea Carol Anne; Dempsey Eugene; Ryan C AnthonyMicrobiome (2017), 5 (1), 4 ISSN:.BACKGROUND: The gut is the most extensively studied niche of the human microbiome. The aim of this study was to characterise the initial gut microbiota development of a cohort of breastfed infants (n = 192) from 1 to 24 weeks of age. METHODS: V4-V5 region 16S rRNA amplicon Illumina sequencing and, in parallel, bacteriological culture. The metabolomic profile of infant urine at 4 weeks of age was also examined by LC-MS. RESULTS: Full-term (FT), spontaneous vaginally delivered (SVD) infants' microbiota remained stable at both phylum and genus levels during the 24-week period examined. FT Caesarean section (CS) infants displayed an increased faecal abundance of Firmicutes (p < 0.01) and lower abundance of Actinobacteria (p < 0.001) after the first week of life compared to FT-SVD infants. FT-CS infants gradually progressed to harbouring a microbiota closely resembling FT-SVD (which remained stable) by week 8 of life, which was maintained at week 24. The gut microbiota of preterm (PT) infants displayed a significantly greater abundance of Proteobacteria compared to FT infants (p < 0.001) at week 1. Metabolomic analysis of urine at week 4 indicated PT-CS infants have a functionally different metabolite profile than FT (both CS and SVD) infants. Co-inertia analysis showed co-variation between the urine metabolome and the faecal microbiota of the infants. Tryptophan and tyrosine metabolic pathways, as well as fatty acid and bile acid metabolism, were found to be affected by delivery mode and gestational age. CONCLUSIONS: These findings confirm that mode of delivery and gestational age both have significant effects on early neonatal microbiota composition. There is also a significant difference between the metabolite profile of FT and PT infants. Prolonged breastfeeding was shown to have a significant effect on the microbiota composition of FT-CS infants at 24 weeks of age, but interestingly not on that of FT-SVD infants. Twins had more similar microbiota to one another than between two random infants, reflecting the influence of similarities in both host genetics and the environment on the microbiota..
- 46Vatanen, T.; Jabbar, K. S.; Ruohtula, T.; Honkanen, J.; Avila-Pacheco, J.; Siljander, H.; Stražar, M.; Oikarinen, S.; Hyöty, H.; Ilonen, J. Mobile genetic elements from the maternal microbiome shape infant gut microbial assembly and metabolism. Cell 2022, 185, 4921– 4936.e4915, DOI: 10.1016/j.cell.2022.11.023Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XjtFOktr7P&md5=1185b098b457b0ef3cfe3ea2f2d498c0Mobile genetic elements from the maternal microbiome shape infant gut microbial assembly and metabolismVatanen, Tommi; Jabbar, Karolina S.; Ruohtula, Terhi; Honkanen, Jarno; Avila-Pacheco, Julian; Siljander, Heli; Strazar, Martin; Oikarinen, Sami; Hyoty, Heikki; Ilonen, Jorma; Mitchell, Caroline M.; Yassour, Moran; Virtanen, Suvi M.; Clish, Clary B.; Plichta, Damian R.; Vlamakis, Hera; Knip, Mikael; Xavier, Ramnik J.Cell (Cambridge, MA, United States) (2022), 185 (26), 4921-4936.e15CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The perinatal period represents a crit. window for cognitive and immune system development, promoted by maternal and infant gut microbiomes and their metabolites. Here, we tracked the co-development of microbiomes and metabolomes from late pregnancy to 1 yr of age using longitudinal multi-omics data from a cohort of 70 mother-infant dyads. We discovered large-scale mother-to-infant interspecies transfer of mobile genetic elements, frequently involving genes assocd. with diet-related adaptations. Infant gut metabolomes were less diverse than maternal but featured hundreds of unique metabolites and microbe-metabolite assocns. not detected in mothers. Metabolomes and serum cytokine signatures of infants who received regular-but not extensively hydrolyzed-formula were distinct from those of exclusively breastfed infants. Taken together, our integrative anal. expands the concept of vertical transmission of the gut microbiome and provides original insights into the development of maternal and infant microbiomes and metabolomes during late pregnancy and early life.
- 47Baleato, C. L.; Ferguson, J. J. A.; Oldmeadow, C.; Mishra, G. D.; Garg, M. L. Plant-Based Dietary Patterns versus Meat Consumption and Prevalence of Impaired Glucose Intolerance and Diabetes Mellitus: A Cross-Sectional Study in Australian Women. Nutrients 2022, 14, 4152, DOI: 10.3390/nu14194152Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xis1yltr%252FJ&md5=3ba139d4a14dc5178154790ac8c366b9Plant-Based Dietary Patterns versus Meat Consumption and Prevalence of Impaired Glucose Intolerance and Diabetes Mellitus: A Cross-Sectional Study in Australian WomenBaleato, Courtney L.; Ferguson, Jessica J. A.; Oldmeadow, Christopher; Mishra, Gita D.; Garg, Manohar L.Nutrients (2022), 14 (19), 4152CODEN: NUTRHU; ISSN:2072-6643. (MDPI AG)This study aimed to compare the prevalence of impaired glucose tolerance (IGT) and diabetes mellitus (DM) among Australian women following plant-based diets (PBD) compared to regular meat eaters. A cross sectional anal. of the mid-aged cohort (1946-1951) of the Australian Longitudinal Study on Women's Health was conducted on completers of Survey 7 in 2013 with complete FFQ data available (n = 9102). Dietary patterns were categorized as PBD (vegan, lacto-ovo vegetarian, pesco-vegetarian, semi-vegetarian) and regular meat eaters. Meat eaters were further categorized into high and low consumption and outcomes included self-reported prevalence of IGT and DM. Participants were identified as regular meat eaters (n = 8937) and PBD (n = 175). Prevalence of IGT was lower in PBD (0-1.2%) compared to regular meat eaters (9.1%). Consolidation of PBD to a single group (vegetarians) indicated a lower prevalence of DM in vegetarians compared to regular meat eaters (3.9% vs. 9.1%). Women consuming meat daily/multiple times per day had significantly higher odds of IGT (OR 1.5, 95%CI 1.1 to 2.1, p = 0.02). Individuals consuming processed meat daily/multiple times per day had significantly higher odds of DM compared to those consuming less than daily (Odds ratio (OR) 1.7, 95% confidence interval (CI) 1.3 to 2.3, p < 0.0001). After adjustment for covariates, statistical significance was lost largely due to the addn. of BMI to the model. Prevalence of IGT and DM were lower in women following PBD and higher in high consumers of meat and processed meat. The relationship between meat consumption and IGT/diabetes status appears to be mediated, at least in part, by an increase in body mass index (BMI). Future studies are warranted to investigate the mechanisms and other lifestyle factors underpinning the assocn. between high meat consumption and increased risk of IGT and DM.
- 48Keaver, L.; Ruan, M.; Chen, F.; Du, M.; Ding, C.; Wang, J.; Shan, Z.; Liu, J.; Zhang, F. F. Plant- and animal-based diet quality and mortality among US adults: a cohort study. Br. J. Nutr. 2021, 125, 1405– 1415, DOI: 10.1017/S0007114520003670Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtFaisLbF&md5=8f55f34569e6b7dd7575a1a841dffd7dPlant- and animal-based diet quality and mortality among US adults: a cohort studyKeaver, Laura; Ruan, Mengyuan; Chen, Fan; Du, Mengxi; Ding, Chenyueyi; Wang, Jiaqi; Shan, Zhilei; Liu, Junxiu; Zhang, Fang FangBritish Journal of Nutrition (2021), 125 (12), 1405-1415CODEN: BJNUAV; ISSN:0007-1145. (Cambridge University Press)Not all plant-based and animal foods exert the same health effects due to their various nutrient compns. We aimed to assess the quality of plant-based v. animal foods in relation to mortality in a prospective cohort study. Using data collected from a nationally representative sample of 36 825 adults in the National Health and Nutrition Examn. Survey 1999-2014, we developed a de novo Comprehensive Diet Quality Index (cDQI) that assesses the quality of seventeen foods based on the healthfulness and sep. scored the quality of eleven plant-based foods in a plant-based Diet Quality Index (pDQI) and six animal foods in an animal-based Diet Quality Index (aDQI). Mortality from all causes, heart disease and cancer were obtained from linkage to the National Death Index up to 31 Dec. 2015. Cox proportional hazard models were used to est. hazard ratios (HR) and 95 % CI after multivariable adjustments. During a median follow-up of 8·3 years, 4669 all-cause deaths occurred, including 798 deaths due to heart disease and 1021 due to cancer. Compared with individuals in the lowest quartile, those in the highest quartile of cDQI had a lower risk of all-cause mortality (HR 0·75, 95 % CI 0·65, 0·86; P trend < 0·001), which largely reflected the inverse relationship between quality of plant-based foods (pDQI) and all-cause mortality (HR 0·66, 95 % CI 0·58, 0·74; P trend < 0·001). No independent assocn. was found for the quality of animal foods (aDQI) and mortality. Our results suggest that consuming healthy plant-based foods is assocd. with lower all-cause mortality among US adults.
- 49O’Mahony, S. M.; Marchesi, J. R.; Scully, P.; Codling, C.; Ceolho, A.-M.; Quigley, E. M. M.; Cryan, J. F.; Dinan, T. G. Early Life Stress Alters Behavior, Immunity, and Microbiota in Rats: Implications for Irritable Bowel Syndrome and Psychiatric Illnesses. Biol. Psychiatry 2009, 65, 263– 267, DOI: 10.1016/j.biopsych.2008.06.026Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1M%252FntlSrug%253D%253D&md5=5fc242a46520953c886d1a25e854f9a0Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnessesO'Mahony Siobhain M; Marchesi Julian R; Scully Paul; Codling Caroline; Ceolho Anne-Marie; Quigley Eamonn M M; Cryan John F; Dinan Timothy GBiological psychiatry (2009), 65 (3), 263-7 ISSN:.BACKGROUND: Adverse early life events are associated with a maladaptive stress response system and might increase the vulnerability to disease in later life. Several disorders have been associated with early life stress, ranging from depression to irritable bowel syndrome. This makes the identification of the neurobiological substrates that are affected by adverse experiences in early life invaluable. METHODS: The purpose of this study was to assess the effect of early life stress on the brain-gut axis. Male rat pups were stressed by separating them from their mothers for 3 hours daily between postnatal days 2-12. The control group was left undisturbed with their mothers. Behavior, immune response, stress sensitivity, visceral sensation, and fecal microbiota were analyzed. RESULTS: The early life stress increased the number of fecal boli in response to a novel stress. Plasma corticosterone was increased in the maternally separated animals. An increase in the systemic immune response was noted in the stressed animals after an in vitro lipopolysaccharide challenge. Increased visceral sensation was seen in the stressed group. There was an alteration of the fecal microbiota when compared with the control group. CONCLUSIONS: These results show that this form of early life stress results in an altered brain-gut axis and is therefore an important model for investigating potential mechanistic insights into stress-related disorders including depression and IBS.
- 50Jernberg, C.; Löfmark, S.; Edlund, C.; Jansson, J. K. Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology (Reading) 2010, 156, 3216– 3223, DOI: 10.1099/mic.0.040618-0Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFCiur3F&md5=56649fa26b86128690cb19944423b1adLong-term impacts of antibiotic exposure on the human intestinal microbiotaJernberg, Cecilia; Loefmark, Sonja; Edlund, Charlotta; Jansson, Janet K.Microbiology (Reading, United Kingdom) (2010), 156 (11), 3216-3223CODEN: MROBEO; ISSN:1350-0872. (Society for General Microbiology)A review. Although it is known that antibiotics have short-term impacts on the human microbiome, recent evidence demonstrates that the impacts of some antibiotics remain for extended periods of time. In addn., antibiotic-resistant strains can persist in the human host environment in the absence of selective pressure. Both mol.- and cultivation-based approaches have revealed ecol. disturbances in the microbiota after antibiotic administration, in particular for specific members of the bacterial community that are susceptible or alternatively resistant to the antibiotic in question. A disturbing consequence of antibiotic treatment has been the long-term persistence of antibiotic resistance genes, for example in the human gut. These data warrant use of prudence in the administration of antibiotics that could aggravate the growing battle with emerging antibiotic-resistant pathogenic strains.
- 51Caporaso, J. G.; Kuczynski, J.; Stombaugh, J.; Bittinger, K.; Bushman, F. D.; Costello, E. K.; Fierer, N.; Peña, A. G.; Goodrich, J. K.; Gordon, J. I. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 2010, 7, 335– 336, DOI: 10.1038/nmeth.f.303Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFalurg%253D&md5=0f3e337921b1cc48bc697f0ee92bf5b8QIIME allows analysis of high-throughput community sequencing dataCaporaso, J. Gregory; Kuczynski, Justin; Stombaugh, Jesse; Bittinger, Kyle; Bushman, Frederic D.; Costello, Elizabeth K.; Fierer, Noah; Pena, Antonio Gonzalez; Goodrich, Julia K.; Gordon, Jeffrey I.; Huttley, Gavin A.; Kelley, Scott T.; Knights, Dan; Koenig, Jeremy E.; Ley, Ruth E.; Lozupone, Catherine A.; McDonald, Daniel; Muegge, Brian D.; Pirrung, Meg; Reeder, Jens; Sevinsky, Joel R.; Turnbaugh, Peter J.; Walters, William A.; Widmann, Jeremy; Yatsunenko, Tanya; Zaneveld, Jesse; Knight, RobNature Methods (2010), 7 (5), 335-336CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)There is no expanded citation for this reference.
- 52Tang, L. Sequence-based identification of human-associated microbiota. https://www.nature.com/articles/d42859-019-00011-5 (accessed October 24, 2022).Google ScholarThere is no corresponding record for this reference.
- 53Collins, S. M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735– 742, DOI: 10.1038/nrmicro2876Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhtlylt7%252FJ&md5=097611d85e63be4c96cb92f83be0367dThe interplay between the intestinal microbiota and the brainCollins, Stephen M.; Surette, Michael; Bercik, PremyslNature Reviews Microbiology (2012), 10 (11), 735-742CODEN: NRMACK; ISSN:1740-1526. (Nature Publishing Group)A review. The intestinal microbiota consists of a vast bacterial community that resides primarily in the lower gut and lives in a symbiotic relationship with the host. A bidirectional neurohumoral communication system, known as the gut-brain axis, integrates the host gut and brain activities. Here, we describe the recent advances in our understanding of how the intestinal microbiota communicates with the brain via this axis to influence brain development and behavior. We also review how this extended communication system might influence a broad spectrum of diseases, including irritable bowel syndrome, psychiatric disorders and demyelinating conditions such as multiple sclerosis.
- 54Lagier, J.-C.; Armougom, F.; Million, M.; Hugon, P.; Pagnier, I.; Robert, C.; Bittar, F.; Fournous, G.; Gimenez, G.; Maraninchi, M. Microbial culturomics: paradigm shift in the human gut microbiome study. Clinical Microbiology and Infection 2012, 18, 1185– 1193, DOI: 10.1111/1469-0691.12023Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1Gru7bP&md5=e7f57683de2adea0585015247a6dd5feMicrobial culturomics: paradigm shift in the human gut microbiome studyLagier, J.-C.; Armougom, F.; Million, M.; Hugon, P.; Pagnier, I.; Robert, C.; Bittar, F.; Fournous, G.; Gimenez, G.; Maraninchi, M.; Trape, J.-F.; Koonin, E. V.; La Scola, B.; Raoult, D.Clinical Microbiology and Infection (2012), 18 (12), 1185-1193CODEN: CMINFM; ISSN:1198-743X. (Wiley-Blackwell)Abstr. : Comprehensive detn. of the microbial compn. of the gut microbiota and the relationships with health and disease are major challenges in the 21st century. Metagenomic anal. of the human gut microbiota detects mostly uncultured bacteria. We studied stools from two lean Africans and one obese European, using 212 different culture conditions (microbial culturomics), and tested the colonies by using mass spectrometry and 16S rRNA amplification and sequencing. In parallel, we analyzed the same three samples by pyrosequencing 16S rRNA amplicons targeting the V6 region. The 32 500 colonies obtained by culturomics have yielded 340 species of bacteria from seven phyla and 117 genera, including two species from rare phyla (Deinococcus-Thermus and Synergistetes), five fungi, and a giant virus (Senegalvirus). The microbiome identified by culturomics included 174 species never described previously in the human gut, including 31 new species and genera for which the genomes were sequenced, generating c. 10 000 new unknown genes (ORFans), which will help in future mol. studies. Among these, the new species Microvirga massiliensis has the largest bacterial genome so far obtained from a human, and Senegalvirus is the largest virus reported in the human gut. Concurrent metagenomic anal. of the same samples produced 698 phylotypes, including 282 known species, 51 of which overlapped with the microbiome identified by culturomics. Thus, culturomics complements metagenomics by overcoming the depth bias inherent in metagenomic approaches.
- 55Smith, P. M.; Howitt, M. R.; Panikov, N.; Michaud, M.; Gallini, C. A.; Bohlooly, Y. M.; Glickman, J. N.; Garrett, W. S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569– 573, DOI: 10.1126/science.1241165Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFyjsr3P&md5=8d7d91434f35758f764d89ab48527cacThe Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell HomeostasisSmith, Patrick M.; Howitt, Michael R.; Panikov, Nicolai; Michaud, Monia; Gallini, Carey Ann; Bohlooly-Y, Mohammad; Glickman, Jonathan N.; Garrett, Wendy S.Science (Washington, DC, United States) (2013), 341 (6145), 569-573CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Regulatory T cells (Tregs) that express the transcription factor Foxp3 are crit. for regulating intestinal inflammation. Candidate microbe approaches have identified bacterial species and strain-specific mols. that can affect intestinal immune responses, including species that modulate Treg responses. Because neither all humans nor mice harbor the same bacterial strains, the authors posited that more prevalent factors exist that regulate the no. and function of colonic Tregs. The authors detd. that short-chain fatty acids, gut microbiota-derived bacterial fermn. products, regulate the size and function of the colonic Treg pool and protect against colitis in a Ffar2-dependent manner in mice. This study reveals that a class of abundant microbial metabolites underlies adaptive immune microbiota coadaptation and promotes colonic homeostasis and health.
- 56Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232– 236, DOI: 10.1038/nature12331Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFShsbfJ&md5=ae3e310120459e75a80926aca55d4f2fTreg induction by a rationally selected mixture of Clostridia strains from the human microbiotaAtarashi, Koji; Tanoue, Takeshi; Oshima, Kenshiro; Suda, Wataru; Nagano, Yuji; Nishikawa, Hiroyoshi; Fukuda, Shinji; Saito, Takuro; Narushima, Seiko; Hase, Koji; Kim, Sangwan; Fritz, Joelle V.; Wilmes, Paul; Ueha, Satoshi; Matsushima, Kouji; Ohno, Hiroshi; Olle, Bernat; Sakaguchi, Shimon; Taniguchi, Tadatsugu; Morita, Hidetoshi; Hattori, Masahira; Honda, KenyaNature (London, United Kingdom) (2013), 500 (7461), 232-236CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Manipulation of the gut microbiota holds great promise for the treatment of inflammatory and allergic diseases. Although numerous probiotic microorganisms have been identified, there remains a compelling need to discover organisms that elicit more robust therapeutic responses, are compatible with the host, and can affect a specific arm of the host immune system in a well-controlled, physiol. manner. Here the authors use a rational approach to isolate CD4+FOXP3+ regulatory T (Treg)-cell-inducing bacterial strains from the human indigenous microbiota. Starting with a healthy human fecal sample, a sequence of selection steps was applied to obtain mice colonized with human microbiota enriched in Treg-cell-inducing species. From these mice, the authors isolated and selected 17 strains of bacteria on the basis of their high potency in enhancing Treg cell abundance and inducing important anti-inflammatory mols.-including interleukin-10 (IL-) and inducible T-cell co-stimulator (ICOS)-in Treg cells upon inoculation into germ-free mice. Genome sequencing revealed that the 17 strains fall within clusters IV, XIVa and XVIII of Clostridia, which lack prominent toxins and virulence factors. The 17 strains act as a community to provide bacterial antigens and a TGF-β-rich environment to help expansion and differentiation of Treg cells. Oral administration of the combination of 17 strains to adult mice attenuated disease in models of colitis and allergic diarrhoea. Use of the isolated strains may allow for tailored therapeutic manipulation of human immune disorders.
- 57Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J. R.; Pfeffer, K.; Coffer, P. J. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451– 455, DOI: 10.1038/nature12726Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFOmtrrJ&md5=5df9a8c09b9b0e7f98522dfe16ef2711Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generationArpaia, Nicholas; Campbell, Clarissa; Fan, Xiying; Dikiy, Stanislav; van der Veeken, Joris; de Roos, Paul; Liu, Hui; Cross, Justin R.; Pfeffer, Klaus; Coffer, Paul J.; Rudensky, Alexander Y.Nature (London, United Kingdom) (2013), 504 (7480), 451-455CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Intestinal microbes provide multicellular hosts with nutrients and confer resistance to infection. The delicate balance between pro- and anti-inflammatory mechanisms, essential for gut immune homeostasis, is affected by the compn. of the commensal microbial community. Regulatory T cells (Treg cells) expressing transcription factor Foxp3 have a key role in limiting inflammatory responses in the intestine. Although specific members of the commensal microbial community have been found to potentiate the generation of anti-inflammatory Treg or pro-inflammatory T helper 17 (TH17) cells, the mol. cues driving this process remain elusive. Considering the vital metabolic function afforded by commensal microorganisms, the authors reasoned that their metabolic byproducts are sensed by cells of the immune system and affect the balance between pro- and anti-inflammatory cells. The authors tested this hypothesis by exploring the effect of microbial metabolites on the generation of anti-inflammatory Treg cells. The authors found that in mice a short-chain fatty acid (SCFA), butyrate, produced by commensal microorganisms during starch fermn., facilitated extrathymic generation of Treg cells. A boost in Treg-cell nos. after provision of butyrate was due to potentiation of extrathymic differentiation of Treg cells, as the obsd. phenomenon was dependent on intronic enhancer CNS1 (conserved non-coding sequence 1), essential for extrathymic but dispensable for thymic Treg-cell differentiation. In addn. to butyrate, de novo Treg-cell generation in the periphery was potentiated by propionate, another SCFA of microbial origin capable of histone deacetylase (HDAC) inhibition, but not acetate, which lacks this HDAC-inhibitory activity. The results suggest that bacterial metabolites mediate communication between the commensal microbiota and the immune system, affecting the balance between pro- and anti-inflammatory mechanisms.
- 58Donia, M. S.; Cimermancic, P.; Schulze, C. J.; Wieland Brown, L. C.; Martin, J.; Mitreva, M.; Clardy, J.; Linington, R. G.; Fischbach, M. A. A systematic analysis of biosynthetic gene clusters in the human microbiome reveals a common family of antibiotics. Cell 2014, 158, 1402– 1414, DOI: 10.1016/j.cell.2014.08.032Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsFCgtLvP&md5=2588272748281d6e3f18966e2c74a702A systematic analysis of biosynthetic gene clusters in the human microbiome reveals a common family of antibioticsDonia, Mohamed S.; Cimermancic, Peter; Schulze, Christopher J.; Wieland Brown, Laura C.; Martin, John; Mitreva, Makedonka; Clardy, Jon; Linington, Roger G.; Fischbach, Michael A.Cell (Cambridge, MA, United States) (2014), 158 (6), 1402-1414CODEN: CELLB5; ISSN:0092-8674. (Cell Press)In complex biol. systems, small mols. often mediate microbe-microbe and microbe-host interactions. Using a systematic approach, the authors identified 3118 small-mol. biosynthetic gene clusters (BGCs) in genomes of human-assocd. bacteria and studied their representation in 752 metagenomic samples from the NIH Human Microbiome Project. Remarkably, they discovered that BGCs for a class of antibiotics in clin. trials, thiopeptides, are widely distributed in genomes and metagenomes of the human microbiota. The authors purified and solved the structure of a thiopeptide antibiotic, lactocillin, from a prominent member of the vaginal microbiota. They demonstrated that lactocillin has potent antibacterial activity against a range of Gram-pos. vaginal pathogens, and they showed that lactocillin and other thiopeptide BGCs are expressed in vivo by analyzing human metatranscriptomic sequencing data. These findings illustrate the widespread distribution of small-mol.-encoding BGCs in the human microbiome, and they demonstrate the bacterial prodn. of drug-like mols. in humans.
- 59Lurie, I.; Yang, Y. X.; Haynes, K.; Mamtani, R.; Boursi, B. Antibiotic exposure and the risk for depression, anxiety, or psychosis: a nested case-control study. J. Clin. Psychiatry 2015, 76, 1522– 1528, DOI: 10.4088/JCP.15m09961Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28visl2isg%253D%253D&md5=2d27297be935450c1c6fd82b0c0d25deAntibiotic exposure and the risk for depression, anxiety, or psychosis: a nested case-control studyLurie Ido; Lurie Ido; Yang Yu-Xiao; Haynes Kevin; Mamtani Ronac; Boursi BenThe Journal of clinical psychiatry (2015), 76 (11), 1522-8 ISSN:.OBJECTIVE: Changes in the microbiota (dysbiosis) were suggested to increase the risk of several psychiatric conditions through neurologic, metabolic, and immunologic pathways. Our aim was to assess whether exposure to specific antibiotic groups increases the risk for depression, anxiety, or psychosis. METHOD: We conducted 3 nested case-control studies during the years 1995-2013 using a large population-based medical record database from the United Kingdom. The study included 202,974 patients with depression, 14,570 with anxiety, and 2,690 with psychosis and 803,961, 57,862, and 10,644 matched controls, respectively. Cases were defined as individuals aged 15-65 years with any medical Read code for depression, anxiety, or psychosis. Subjects with diagnosis-specific psychotropic prescriptions > 90 days before index date were excluded. For every case, 4 controls were selected using incidence density sampling, matching on age, sex, practice site, calendar time, and duration of follow-up before index date. The primary exposure of interest was therapy with 1 of 7 antibiotic classes > 1 year before index date. Odds ratios (ORs) and 95% CIs were calculated for the association between each psychiatric disorder and exposure to individual classes of antibiotics using conditional logistic regression analysis. The risk was adjusted for obesity, smoking history, alcohol consumption, socioeconomic status, and number of infectious events before diagnosis. RESULTS: Treatment with a single antibiotic course was associated with higher risk for depression with all antibiotic groups, with an adjusted OR (AOR) of 1.23 for penicillins (95% CI, 1.18-1.29) and 1.25 (95% CI, 1.15-1.35) for quinolones. The risk increased with recurrent antibiotic exposures to 1.40 (95% CI, 1.35-1.46) and 1.56 (95% CI, 1.46-1.65) for 2-5 and > 5 courses of penicillin, respectively. Similar association was observed for anxiety and was most prominent with exposures to penicillins and sulfonamides, with an AOR of 1.17 (95% CI, 1.01-1.36) for a single course of penicillin and 1.44 (95% CI, 1.18-1.75) for > 5 courses. There was no change in risk for psychosis with any antibiotic group. There was a mild increase in the risk of depression and anxiety with a single course of antifungals; however, there was no increase in risk with repeated exposures. CONCLUSION: Recurrent antibiotic exposure is associated with increased risk for depression and anxiety but not for psychosis.
- 60Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.; Fan, S.; Du, X. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatry 2016, 21, 786– 796, DOI: 10.1038/mp.2016.44Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmsVals78%253D&md5=58e19b9beef57cc353c806891f3ef2f0Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host's metabolismZheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.; Fan, S.; Du, X.; Zhang, X.; Yang, D.; Yang, Y.; Meng, H.; Li, W.; Melgiri, N. D.; Licinio, J.; Wei, H.; Xie, P.Molecular Psychiatry (2016), 21 (6), 786-796CODEN: MOPSFQ; ISSN:1359-4184. (Nature Publishing Group)Major depressive disorder (MDD) is the result of complex gene-environment interactions. According to the World Health Organization, MDD is the leading cause of disability worldwide, and it is a major contributor to the overall global burden of disease. However, the definitive environmental mechanisms underlying the pathophysiol. of MDD remain elusive. The gut microbiome is an increasingly recognized environmental factor that can shape the brain through the microbiota-gut-brain axis. We show here that the absence of gut microbiota in germ-free (GF) mice resulted in decreased immobility time in the forced swimming test relative to conventionally raised healthy control mice. Moreover, from clin. sampling, the gut microbiotic compns. of MDD patients and healthy controls were significantly different with MDD patients characterized by significant changes in the relative abundance of Firmicutes, Actinobacteria and Bacteroidetes. Fecal microbiota transplantation of GF mice with 'depression microbiota' derived from MDD patients resulted in depression-like behaviors compared with colonization with 'healthy microbiota' derived from healthy control individuals. Mice harboring 'depression microbiota' primarily exhibited disturbances of microbial genes and host metabolites involved in carbohydrate and amino acid metab. This study demonstrates that dysbiosis of the gut microbiome may have a causal role in the development of depressive-like behaviors, in a pathway that is mediated through the host's metab.
- 61Davis, D. J.; Hecht, P. M.; Jasarevic, E.; Beversdorf, D. Q.; Will, M. J.; Fritsche, K.; Gillespie, C. H. Sex-specific effects of docosahexaenoic acid (DHA) on the microbiome and behavior of socially-isolated mice. Brain, Behavior, and Immunity 2017, 59, 38– 48, DOI: 10.1016/j.bbi.2016.09.003Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsV2hu7bF&md5=f827c7e72227a384d1ab9d225bc3b2beSex-specific effects of docosahexaenoic acid (DHA) on the microbiome and behavior of socially-isolated miceDavis, Daniel J.; Hecht, Patrick M.; Jasarevic, Eldin; Beversdorf, David Q.; Will, Matthew J.; Fritsche, Kevin; Gillespie, Catherine H.Brain, Behavior, and Immunity (2017), 59 (), 38-48CODEN: BBIMEW; ISSN:0889-1591. (Elsevier)Dietary supplementation with the long-chain omega-3 polyunsatd. fatty acid docosahexaenoic acid (DHA) has been shown to have a beneficial effect on reducing the symptoms assocd. with several neuropsychiatric conditions including anxiety and depression. However, the mechanisms underlying this effect remain largely unknown. Increasing evidence suggests that the vast repertoire of commensal bacteria within the gut plays a crit. role in regulating various biol. processes in the brain and may contribute to neuropsychiatric disease risk. The present study detd. the contribution of DHA on anxiety and depressive-like behaviors through modulation of the gut microbiota in a paradigm of social isolation. Adult male and female mice were subjected to social isolation for 28 days and then placed either on a control diet or a diet supplemented with 0.1% or 1.0% DHA. Fecal pellets were collected both 24 h and 7 days following the introduction of the new diets. Behavioral testing revealed that male mice fed a DHA diet, regardless of dose, exhibited reduced anxiety and depressive-like behaviors compared to control fed mice while no differences were obsd. in female mice. As the microbiota-brain-axis has been recently implicated in behavior, compn. of microbial communities were analyzed to examine if these sex-specific effects of DHA may be assocd. with changes in the gut microbiota (GM). Clear sex differences were obsd. with males and females showing distinct microbial compns. prior to DHA supplementation. The introduction of DHA into the diet also induced sex-specific interactions on the GM with the fatty acid producing a significant effect on the microbial profiles in males but not in females. Interestingly, levels of Allobaculum and Ruminococcus were found to significantly correlate with the behavioral changes obsd. in the male mice. Predictive metagenome anal. using PICRUSt was performed on the fecal samples collected from males and identified enrichment in functional KEGG pathway terms relevant to processes such as the biosynthesis of unsatd. fatty acids and antioxidant metab. These results indicate that DHA alters commensal community compn. and produces beneficial effects on anxiety and depressive-like behaviors in a sex-specific manner. The present study provides insight into the mechanistic role that gut microbes may play in the regulation of anxiety and depressive-like behaviors and how dietary intervention can modulate these effects.
- 62Pasolli, E.; Asnicar, F.; Manara, S.; Zolfo, M.; Karcher, N.; Armanini, F.; Beghini, F.; Manghi, P.; Tett, A.; Ghensi, P. Extensive Unexplored Human Microbiome Diversity Revealed by Over 150,000 Genomes from Metagenomes Spanning Age, Geography, and Lifestyle. Cell 2019, 176, 649– 662.e20, DOI: 10.1016/j.cell.2019.01.001Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsVars7w%253D&md5=28875385d40b31967e02ad1538cf2c57Extensive Unexplored Human Microbiome Diversity Revealed by Over 150,000 Genomes from Metagenomes Spanning Age, Geography, and LifestylePasolli, Edoardo; Asnicar, Francesco; Manara, Serena; Zolfo, Moreno; Karcher, Nicolai; Armanini, Federica; Beghini, Francesco; Manghi, Paolo; Tett, Adrian; Ghensi, Paolo; Collado, Maria Carmen; Rice, Benjamin L.; DuLong, Casey; Morgan, Xochitl C.; Golden, Christopher D.; Quince, Christopher; Huttenhower, Curtis; Segata, NicolaCell (Cambridge, MA, United States) (2019), 176 (3), 649-662.e20CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The body-wide human microbiome plays a role in health, but its full diversity remains uncharacterized, particularly outside of the gut and in international populations. We leveraged 9,428 metagenomes to reconstruct 154,723 microbial genomes (45% of high quality) spanning body sites, ages, countries, and lifestyles. We recapitulated 4,930 species-level genome bins (SGBs), 77% without genomes in public repositories (unknown SGBs [uSGBs]). uSGBs are prevalent (in 93% of well-assembled samples), expand underrepresented phyla, and are enriched in non-Westernized populations (40% of the total SGBs). We annotated 2.85 M genes in SGBs, many assocd. with conditions including infant development (94,000) or Westernization (106,000). SGBs and uSGBs permit deeper microbiome analyses and increase the av. mappability of metagenomic reads from 67.76% to 87.51% in the gut (median 94.26%) and 65.14% to 82.34% in the mouth. We thus identify thousands of microbial genomes from yet-to-be-named species, expand the pangenomes of human-assocd. microbes, and allow better exploitation of metagenomic technologies.
- 63Nayfach, S.; Shi, Z. J.; Seshadri, R.; Pollard, K. S.; Kyrpides, N. C. New insights from uncultivated genomes of the global human gut microbiome. Nature 2019, 568, 505– 510, DOI: 10.1038/s41586-019-1058-xGoogle Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXosVSgsbo%253D&md5=3a9d99af65b1dfad3b0e591fa19c8e16New insights from uncultivated genomes of the global human gut microbiomeNayfach, Stephen; Shi, Zhou Jason; Seshadri, Rekha; Pollard, Katherine S.; Kyrpides, Nikos C.Nature (London, United Kingdom) (2019), 568 (7753), 505-510CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The genome sequences of many species of the human gut microbiome remain unknown, largely owing to challenges in cultivating microorganisms under lab. conditions. We address this problem by reconstructing 60,664 draft prokaryotic genomes from 3810 fecal metagenomes, from geog. and phenotypically diverse humans. These genomes provide ref. points for 2058 newly identified species-level operational taxonomic units (OTUs), which represents a 50% increase over the previously known phylogenetic diversity of sequenced gut bacteria. On av., the newly identified OTUs comprise 33% of richness and 28% of species abundance per individual, and are enriched in humans from rural populations. A meta-anal. of clin. gut-microbiome studies pinpointed numerous disease assocns. for the newly identified OTUs, which have the potential to improve predictive models. Finally, our anal. revealed that uncultured gut species have undergone genome redn. that has resulted in the loss of certain biosynthetic pathways, which may offer clues for improving cultivation strategies in the future.
- 64Pusceddu, M. M.; Barboza, M.; Schneider, M.; Stokes, P.; Sladek, J. A.; Torres-Fuentes, C.; Goldfild, L. R.; Gillis, S. E.; Brust-Mascher, I.; Rabasa, G. Nod-like receptors are critical for gut-brain axis signaling in mice. J. Physiol. 2019, 597, 5777– 5797, DOI: 10.1113/JP278640Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1arurzK&md5=a061ded6626d4888d224db082722f1d2Nod-like receptors are critical for gut-brain axis signalling in micePusceddu, Matteo M.; Barboza, Mariana; Keogh, Ciara E.; Schneider, Melinda; Stokes, Patricia; Sladek, Jessica A.; Kim, Hyun Jung D.; Torres-Fuentes, Cristina; Goldfild, Lily R.; Gillis, Shane E.; Brust-Mascher, Ingrid; Rabasa, Gonzalo; Wong, Kyle A.; Lebrilla, Carlito; Byndloss, Mariana X.; Maisonneuve, Charles; Baeumler, Andreas J.; Philpott, Dana J.; Ferrero, Richard L.; Barrett, Kim E.; Reardon, Colin; Gareau, Melanie G.Journal of Physiology (Oxford, United Kingdom) (2019), 597 (24), 5777-5797CODEN: JPHYA7; ISSN:1469-7793. (Wiley-Blackwell)Key points : ·Nucleotide binding oligomerization domain (Nod)-like receptors regulate cognition, anxiety and hypothalamic-pituitary-adrenal axis activation. ·Nod-like receptors regulate central and peripheral serotonergic biol. ·Nod-like receptors are important for maintenance of gastrointestinal physiol. ·Intestinal epithelial cell expression of Nod1 receptors regulate behavior. Gut-brain axis signalling is crit. for maintaining health and homeostasis. Stressful life events can impact gut-brain signalling, leading to altered mood, cognition and intestinal dysfunction. In the present study, we identified nucleotide binding oligomerization domain (Nod)-like receptors (NLR), Nod1 and Nod2, as novel regulators for gut-brain signalling. NLR are innate immune pattern recognition receptors expressed in the gut and brain, and are important in the regulation of gastrointestinal physiol. We found that mice deficient in both Nod1 and Nod2 (NodDKO) demonstrate signs of stress-induced anxiety, cognitive impairment and depression in the context of a hyperactive hypothalamic-pituitary-adrenal axis. These deficits were coupled with impairments in the serotonergic pathway in the brain, decreased hippocampal cell proliferation and immature neurons, as well as reduced neural activation. In addn., NodDKO mice had increased gastrointestinal permeability and altered serotonin signalling in the gut following exposure to acute stress.
- 65Pellegrini, C.; Antonioli, L.; Calderone, V.; Colucci, R.; Fornai, M.; Blandizzi, C. Microbiota-gut-brain axis in health and disease: Is NLRP3 inflammasome at the crossroads of microbiota-gut-brain communications?. Prog. Neurobiol. 2020, 191, 101806, DOI: 10.1016/j.pneurobio.2020.101806Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVClsrrN&md5=81eaecf674dc6963a355a67b47845bb0Microbiota-gut-brain axis in health and disease: Is NLRP3 inflammasome at the crossroads of microbiota-gut-brain communicationsPellegrini, Carolina; Antonioli, Luca; Calderone, Vincenzo; Colucci, Rocchina; Fornai, Matteo; Blandizzi, CorradoProgress in Neurobiology (Oxford, United Kingdom) (2020), 191 (), 101806CODEN: PGNBA5; ISSN:0301-0082. (Elsevier Ltd.)A review. Growing evidence highlights the relevance of microbiota-gut-brain axis in the maintenance of brain homeostasis as well as in the pathophysiol. of major neurol. and psychiatric disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), autism spectrum disorder (ASD) and major depressive disorder (MDD). Of note, the nucleotide-binding oligomerization domain leucine rich repeat and pyrin domain-contg. protein 3 (NLRP3) inflammasome acts as a key player in both coordinating the host physiol. and shaping the peripheral and central immune/inflammatory responses in CNS diseases. The present review provides an overview of current knowledge on the role of microbiota-gut-inflammasome-brain axis in the major CNS diseases, including PD, AD, MS, ASD and MDD. In particular, though no direct and causal correlation among altered gut microbiota, NLRP3 activation and brain pathol. has been demonstrated and in-depth studies are needed in this setting, our purpose was to pave the way to a novel and pioneering perspective on the pathophysiol. of CNS disorders. Our intent was also to highlight and discuss whether alterations of microbiota-gut-inflammasome-brain axis support a holistic view of the pathophysiol. of CNS diseases, even though each disorder displays a different clin. picture.
- 66Zhao, K.; Yao, M.; Zhang, X.; Xu, F.; Shao, X.; Wei, Y.; Wang, H. Flavonoids and intestinal microbes interact to alleviate depression. Journal of the Science of Food and Agriculture 2022, 102, 1311– 1318, DOI: 10.1002/jsfa.11578Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitlCntbvF&md5=53b83d6529ef0b11cacf7b6b687c9fbbFlavonoids and intestinal microbes interact to alleviate depressionZhao, Ke; Yao, Mei; Zhang, Xin; Xu, Feng; Shao, Xingfeng; Wei, Yingying; Wang, HongfeiJournal of the Science of Food and Agriculture (2022), 102 (4), 1311-1318CODEN: JSFAAE; ISSN:0022-5142. (John Wiley & Sons Ltd.)A review. Flavonoids have a variety of biol. activities that are beneficial to human health. However, owing to low bioavailability, most flavonoids exert beneficial effects in the intestine through metab. by the flora into a variety of structurally different derivs. Also, flavonoids can modulate the type and structure of intestinal microorganisms to improve human health. It has been reported that the development of depression is accompanied by changes in the type and no. of intestinal microorganisms, and gut microbes can significantly improve depressive symptoms through the gut-brain axis. Therefore, the interaction between flavonoids and intestinal microbes to alleviate depression is discussed.
- 67Segre, J. A. What does it take to satisfy Koch’s postulates two centuries later? Microbial genomics and Propionibacteria acnes. J. Invest. Dermatol. 2013, 133, 2141– 2142, DOI: 10.1038/jid.2013.260Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVykur%252FP&md5=165251fa065804d5cdc60736bf7e939eWhat Does It Take to Satisfy Koch's Postulates Two Centuries Later?: Microbial Genomics and Propionibacteria acnesSegre, Julia A.Journal of Investigative Dermatology (2013), 133 (9), 2141-2142CODEN: JIDEAE; ISSN:0022-202X. (Nature Publishing Group)For two centuries, Koch's postulates have set the gold std. for establishing the microbiol. etiol. of infection and disease. Genomic sequencing now brings finer resoln. to both bacterial strain variation and the host genetic state that may predispose to disease. In this issue of the JID, Fitz-Gibbons and colleagues present strain-based resoln. of Propionibacterium acnes and its assocn. with the common teenage malady acne vulgaris. Here I examine how Koch's postulates were envisioned and incorporate this finer resoln. of both host and microbial states.
- 68Eiseman, B.; Silen, W.; Bascom, G. S.; Kauvar, A. J. Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitis. Surgery 1958, 44, 854– 859Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaG1M%252Fit1ShtA%253D%253D&md5=932cb97103c858a9f7c3112e2e61dca1Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitisEISEMAN B; SILEN W; BASCOM G S; KAUVAR A JSurgery (1958), 44 (5), 854-9 ISSN:0039-6060.There is no expanded citation for this reference.
- 69Reyniers, J. A. The pure-culture concept and gnotobiotics. Ann. N.Y. Acad. Sci. 1959, 78, 3– 16, DOI: 10.1111/j.1749-6632.1959.tb53091.xGoogle ScholarThere is no corresponding record for this reference.
- 70Schaedler, R. W.; Dubos, R.; Costello, R. ASSOCIATION OF GERMFREE MICE WITH BACTERIA ISOLATED FROM NORMAL MICE. J. Exp. Med. 1965, 122, 77– 82, DOI: 10.1084/jem.122.1.77Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaF2M7jtVWmsQ%253D%253D&md5=c693bd4d999907652054e8ac1c18c68cASSOCIATION OF GERMFREE MICE WITH BACTERIA ISOLATED FROM NORMAL MICESCHAEDLER R W; DUBS R; COSTELLO RThe Journal of experimental medicine (1965), 122 (), 77-82 ISSN:0022-1007.Germfree mice were given food contaminated with pure cultures of various bacterial species isolated from ordinary healthy mice. The cultures were given singly, or in association, or consecutively at weekly intervals. Whatever the technique of administration, the lactobacilli and anaerobic streptococci immediately established themselves throughout the gastrointestinal tract, and became closely associated with the walls of the organs. In contrast, the organisms of the bacteroides group were found in large numbers only in the large intestine. Within a week after exposure, the populations of these three bacterial species reached levels similar to those found in ordinary mice. They remained at these characteristic levels throughout the period of observation (several months). Their presence resulted in a progressive decrease in the size of the cecum which eventually became normal in gross appearance. Coliform bacilli multiplied extensively and persisted at high levels in all parts of the gastrointestinal tract of germfree mice, even after these had become colonized with lactobacilli, anaerobic streptococci and bacteroides. However, the coliform population fell precipitously within a few days after the animals were fed the intestinal contents of healthy pathogen-free mice.
- 71Peppercorn, M. A.; Goldman, P. The Role of Intestinal Bacteria in the Metabolism of Salicylazosulfapyridine. Journal of Pharmacology and Experimental Therapeutics 1972, 181, 555– 562Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE38Xks12qt7c%253D&md5=149105b21c094828504bc77d66df2b80Role of intestinal bacteria in the metabolism of salicylazosulfapyridinePeppercorn, Mark A.; Goldman, PeterJournal of Pharmacology and Experimental Therapeutics (1972), 181 (3), 555-62CODEN: JPETAB; ISSN:0022-3565.Following oral administration of salicylazosulfapyridine (I) [599-79-1] to normal rats, sulfapyridine [144-83-2], 5-aminosalicylate [89-57-6], and their N-acetyl derivs. were found in the excreta. No unchanged I was found. Pretreatment of the rats with neomycin [1404-04-2] in order to decrease intestinal microflora resulted in fecal recovery of >50% of the administered dose of I as unchanged I. Intestinal bacteria appear responsible for the initial reaction in I biotransformation and this raises questions concerning the amt. of unchanged I that reaches the presumed site of action in inflammatory disease of the lower intestine.
- 72Stark, P. L.; Lee, A. The microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of life. J. Med. Microbiol. 1982, 15, 189– 203, DOI: 10.1099/00222615-15-2-189Google Scholar72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL3s%252FlvFGqtw%253D%253D&md5=4c3b7442ffffe9234aebbad8912d627dThe microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of lifeStark P L; Lee AJournal of medical microbiology (1982), 15 (2), 189-203 ISSN:0022-2615.The succession of bacterial populations in the large bowel of seven breast-fed and seven formula-fed infants was examined during the first year of life. The composition of the intestinal microflora varied according to the infant's diet. During the first week of life breast-fed and formula-fed infants were colonised by enterobacteria and enterococci followed by bifidobacteria, Bacteroides spp., clostridia and anaerobic streptococci. From week 4 until solid foods were given, breast-fed babies had a simple flora consisting of bifidobacteria and relatively few enterobacteria and enterococci. Formula-fed babies during the corresponding period were more often colonised by other anaerobes in addition to bifidobacteria and had higher counts of facultatively anaerobic bacteria. The introduction of solid food to the breast-fed infants caused a major disturbance in the microbial ecology of the large bowel as counts of enterobacteria and enterococci rose sharply and colonisation by Bacteroides spp., clostridia and anaerobic streptococci occurred. This was not observed when formula-fed infants began to take solids; instead, counts of facultative anaerobes remained high while colonisation by anaerobes other than bifidobacteria continued. At 12 months, the anaerobic bacterial populations of the large bowel of breast-fed and formula-fed infants were beginning to resemble those of adults in number and composition and there was a corresponding decrease in the number of facultative anaerobes. These changes are discussed in relation to changes in susceptibility to gastro-intestinal infection.
- 73Fleischmann, R. D.; Adams, M. D.; White, O.; Clayton, R. A.; Kirkness, E. F.; Kerlavage, A. R.; Bult, C. J.; Tomb, J. F.; Dougherty, B. A.; Merrick, J. M. Whole-genome random sequencing and assembly of Haemophilus influenzae Rd. Science 1995, 269, 496– 512, DOI: 10.1126/science.7542800Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXntF2ksLc%253D&md5=471dc5b1db156773d4883e41b78ece77Whole-genome random sequencing and assembly of Haemophilus influenzae RdFleischmann, Robert D.; Adams, Mark D.; White, Owen; Clayton, Rebecca A.; Kirkness, Ewen F.; Kerlavage, Anthony R.; Bult, Carol J.; Tomb, Jean-Francois; Dougherty, Brian A.; et al.Science (Washington, D. C.) (1995), 269 (5223), 496-8, 507-12CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)An approach for genome anal. based on sequencing and assembly of unselected pieces of DNA from the whole chromosome has been applied to obtain the complete nucleotide sequence (1,830,137 base pairs) of the genome from the bacterium Haemophilus influenzae Rd. This approach eliminates the need for initial mapping efforts and is therefore applicable to the vast array of microbial species for which genomes maps are unavailable. The H. influenzae Rd genome sequence (Genome Sequence DataBase accession no. L42023) represents the only complete genome sequence from a free-living organism.
- 74Wilson, K. H.; Blitchington, R. B. Human colonic biota studied by ribosomal DNA sequence analysis. Appl. Environ. Microbiol. 1996, 62, 2273– 2278, DOI: 10.1128/aem.62.7.2273-2278.1996Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XjvFGntro%253D&md5=a29cae5d95717df891d1b69cd23dcf93Human colonic biota studied by ribosomal DNA sequence analysisWilson, Kenneth H.; Blitchington, Rhonda B.Applied and Environmental Microbiology (1996), 62 (7), 2273-2278CODEN: AEMIDF; ISSN:0099-2240. (American Society for Microbiology)Human colonic biota is a complex microbial ecosystem that serves as a host defense. Unlike most microbial ecosystems, its compn. has been studied extensively by relatively efficient culture methods. The authors have compared an established culture-based method with direct amplification and partial sequencing of cloned 16S rRNA genes from a human fecal specimen. Nine cycles of PCR were also compared with 35 cycles. Colonies and cloned amplicons were classified by comparing their ribosomal DNA (rDNA; DNA coding for rRNA) sequences with rDNA sequences of known phylogeny. Quant. culture recovered 58% of the microscopic count. The 48 colonies identified gave 21 rDNA sequences; it was estd. that 72% of the rDNA sequences from the total population of culturable cells would match these 21 sampled sequences (72% coverage). Fifty 9-cycle clones gave 27 sequences and 59% coverage of cloned rDNAs. Thirty-nine rDNAs cloned after 35 cycles of PCR gave 13 sequences for 74% coverage. Thus, the representation of the ecosystem after 35 cycles of PCR was distorted and lacked diversity. However, when the no. of temp. cycles was minimized, biodiversity was preserved, and there was good agreement between culturing bacteria and sampling rDNA directly.
- 75Guarner, F.; Malagelada, J. R. Gut flora in health and disease. Lancet 2003, 361, 512– 519, DOI: 10.1016/S0140-6736(03)12489-0Google Scholar75https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3s%252FntlKltg%253D%253D&md5=fd95e325ed4ca919a4d28659eb506038Gut flora in health and diseaseGuarner Francisco; Malagelada Juan-RLancet (London, England) (2003), 361 (9356), 512-9 ISSN:0140-6736.The human gut is the natural habitat for a large and dynamic bacterial community, but a substantial part of these bacterial populations are still to be described. However, the relevance and effect of resident bacteria on a host's physiology and pathology has been well documented. Major functions of the gut microflora include metabolic activities that result in salvage of energy and absorbable nutrients, important trophic effects on intestinal epithelia and on immune structure and function, and protection of the colonised host against invasion by alien microbes. Gut flora might also be an essential factor in certain pathological disorders, including multisystem organ failure, colon cancer, and inflammatory bowel diseases. Nevertheless, bacteria are also useful in promotion of human health. Probiotics and prebiotics are known to have a role in prevention or treatment of some diseases.
- 76Rakoff-Nahoum, S.; Paglino, J.; Eslami-Varzaneh, F.; Edberg, S.; Medzhitov, R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004, 118, 229– 241, DOI: 10.1016/j.cell.2004.07.002Google Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmtlKntrg%253D&md5=08ed04f1969160743c82bae2b70a201bRecognition of commensal microflora by Toll-like receptors is required for intestinal homeostasisRakoff-Nahoum, Seth; Paglino, Justin; Eslami-Varzaneh, Fatima; Edberg, Stephen; Medzhitov, RuslanCell (Cambridge, MA, United States) (2004), 118 (2), 229-241CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Toll-like receptors (TLRs) play a crucial role in host defense against microbial infection. The microbial ligands recognized by TLRs are not unique to pathogens, however, and are produced by both pathogenic and commensal microorganisms. It is thought that an inflammatory response to commensal bacteria is avoided due to sequestration of microflora by surface epithelia. Here, we show that commensal bacteria are recognized by TLRs under normal steady-state conditions, and this interaction plays a crucial role in the maintenance of intestinal epithelial homeostasis. Furthermore, we find that activation of TLRs by commensal microflora is crit. for the protection against gut injury and assocd. mortality. These findings reveal a novel function of TLRs-control of intestinal epithelial homeostasis and protection from injury-and provide a new perspective on the evolution of host-microbial interactions.
- 77Mazmanian, S. K.; Liu, C. H.; Tzianabos, A. O.; Kasper, D. L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005, 122, 107– 118, DOI: 10.1016/j.cell.2005.05.007Google Scholar77https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmsFeiurw%253D&md5=029fa2f2e90305138740c220055e4f15An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune systemMazmanian, Sarkis K.; Liu, Cui Hua; Tzianabos, Arthur O.; Kasper, Dennis L.Cell (Cambridge, MA, United States) (2005), 122 (1), 107-118CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The mammalian gastrointestinal tract harbors a complex ecosystem consisting of countless bacteria in homeostasis with the host immune system. Shaped by evolution, this partnership has potential for symbiotic benefit. However, the identities of bacterial mols. mediating symbiosis remain undefined. Here the authors show that, during colonization of animals with the ubiquitous gut microorganism Bacteroides fragilis, a bacterial polysaccharide (PSA) directs the cellular and phys. maturation of the developing immune system. Comparison with germ-free animals reveals that the immunomodulatory activities of PSA during B. fragilis colonization include correcting systemic T cell deficiencies and TH1/TH2 imbalances and directing lymphoid organogenesis. A PSA mutant of B. fragilis does not restore these immunol. functions. PSA presented by intestinal dendritic cells activates CD4+ T cells and elicits appropriate cytokine prodn. These findings provide a mol. basis for host-bacterial symbiosis and reveal the archetypal mol. of commensal bacteria that mediates development of the host immune system.
- 78Turnbaugh, P. J.; Ley, R. E.; Mahowald, M. A.; Magrini, V.; Mardis, E. R.; Gordon, J. I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027– 1031, DOI: 10.1038/nature05414Google Scholar78https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjslWgurs%253D&md5=628d0c9769232cc726ebf8f2d19068eeAn obesity-associated gut microbiome with increased capacity for energy harvestTurnbaugh, Peter J.; Ley, Ruth E.; Mahowald, Michael A.; Magrini, Vincent; Mardis, Elaine R.; Gordon, Jeffrey I.Nature (London, United Kingdom) (2006), 444 (7122), 1027-1031CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The worldwide obesity epidemic is stimulating efforts to identify host and environmental factors that affect energy balance. Comparisons of the distal gut microbiota of genetically obese mice and their lean littermates, as well as those of obese and lean human volunteers have revealed that obesity is assocd. with changes in the relative abundance of the two dominant bacterial divisions, the Bacteroidetes and the Firmicutes. Here we demonstrate through meta-genomic and biochem. analyses that these changes affect the metabolic potential of the mouse gut microbiota. Our results indicate that the obese microbiome has an increased capacity to harvest energy from the diet. Furthermore, this trait is transmissible: colonization of germ-free mice with an 'obese microbiota' results in a significantly greater increase in total body fat than colonization with a 'lean microbiota'. These results identify the gut microbiota as an addnl. contributing factor to the pathophysiol. of obesity.
- 79Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C. P. M.; Alou, M. T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M. P. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 2018, 359, 91– 97, DOI: 10.1126/science.aan3706Google Scholar79https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjslOrsw%253D%253D&md5=a095f1934782cd73323256ab75957462Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumorsRouty, Bertrand; Le Chatelier, Emmanuelle; Derosa, Lisa; Duong, Connie P. M.; Alou, Maryam Tidjani; Daillere, Romain; Fluckiger, Aurelie; Messaoudene, Meriem; Rauber, Conrad; Roberti, Maria P.; Fidelle, Marine; Flament, Caroline; Poirier-Colame, Vichnou; Opolon, Paule; Klein, Christophe; Iribarren, Kristina; Mondragon, Laura; Jacquelot, Nicolas; Qu, Bo; Ferrere, Gladys; Clemenson, Celine; Mezquita, Laura; Masip, Jordi Remon; Naltet, Charles; Brosseau, Solenn; Kaderbhai, Coureche; Richard, Corentin; Rizvi, Hira; Levenez, Florence; Galleron, Nathalie; Quinquis, Benoit; Pons, Nicolas; Ryffel, Bernhard; Minard-Colin, Veronique; Gonin, Patrick; Soria, Jean-Charles; Deutsch, Eric; Loriot, Yohann; Ghiringhelli, Francois; Zalcman, Gerard; Goldwasser, Francois; Escudier, Bernard; Hellmann, Matthew D.; Eggermont, Alexander; Raoult, Didier; Albiges, Laurence; Kroemer, Guido; Zitvogel, LaurenceScience (Washington, DC, United States) (2018), 359 (6371), 91-97CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis induce sustained clin. responses in a sizable minority of cancer patients. We found that primary resistance to ICIs can be attributed to abnormal gut microbiome compn. Antibiotics inhibited the clin. benefit of ICIs in patients with advanced cancer. Fecal microbiota transplantation (FMT) from cancer patients who responded to ICIs into germ-free or antibiotic-treated mice ameliorated the antitumor effects of PD-1 blockade, whereas FMT from nonresponding patients failed to do so. Metagenomics of patient stool samples at diagnosis revealed correlations between clin. responses to ICIs and the relative abundance of Akkermansia muciniphila. Oral supplementation with A. muciniphila after FMT with nonresponder feces restored the efficacy of PD-1 blockade in an interleukin-12-dependent manner by increasing the recruitment of CCR9+CXCR3+CD4+ T lymphocytes into mouse tumor beds.
- 80Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M. L.; Luke, J. J.; Gajewski, T. F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104– 108, DOI: 10.1126/science.aao3290Google Scholar80https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjslOksg%253D%253D&md5=961e9b039d3cdb440d618640ad218dadThe commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patientsMatson, Vyara; Fessler, Jessica; Bao, Riyue; Chongsuwat, Tara; Zha, Yuanyuan; Alegre, Maria-Luisa; Luke, Jason J.; Gajewski, Thomas F.Science (Washington, DC, United States) (2018), 359 (6371), 104-108CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Anti-PD-1-based immunotherapy has had a major impact on cancer treatment but has only benefited a subset of patients. Among the variables that could contribute to interpatient heterogeneity is differential compn. of the patients' microbiome, which has been shown to affect antitumor immunity and immunotherapy efficacy in preclin. mouse models. The authors analyzed baseline stool samples from metastatic melanoma patients before immunotherapy treatment, through an integration of 16S rRNA gene sequencing, metagenomic shotgun sequencing, and quant. polymerase chain reaction for selected bacteria. A significant assocn. was obsd. between commensal microbial compn. and clin. response. Bacterial species more abundant in responders included Bifidobacterium longum, Collinsella aerofaciens, and Enterococcus faecium. Reconstitution of germ-free mice with fecal material from responding patients could lead to improved tumor control, augmented T cell responses, and greater efficacy of anti-PD-L1 therapy. The authors' results suggest that the commensal microbiome may have a mechanistic impact on antitumor immunity in human cancer patients.
- 81Belizário, J. E.; Napolitano, M. Human microbiomes and their roles in dysbiosis, common diseases, and novel therapeutic approaches. Front. Microbiol. 2015, 6, 1050, DOI: 10.3389/fmicb.2015.01050Google Scholar81https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28zlslWmtA%253D%253D&md5=9c50c0d119d45e1f45a9d16073e0eda2Human microbiomes and their roles in dysbiosis, common diseases, and novel therapeutic approachesBelizario Jose E; Napolitano MauroFrontiers in microbiology (2015), 6 (), 1050 ISSN:1664-302X.The human body is the residence of a large number of commensal (non-pathogenic) and pathogenic microbial species that have co-evolved with the human genome, adaptive immune system, and diet. With recent advances in DNA-based technologies, we initiated the exploration of bacterial gene functions and their role in human health. The main goal of the human microbiome project is to characterize the abundance, diversity and functionality of the genes present in all microorganisms that permanently live in different sites of the human body. The gut microbiota expresses over 3.3 million bacterial genes, while the human genome expresses only 20 thousand genes. Microbe gene-products exert pivotal functions via the regulation of food digestion and immune system development. Studies are confirming that manipulation of non-pathogenic bacterial strains in the host can stimulate the recovery of the immune response to pathogenic bacteria causing diseases. Different approaches, including the use of nutraceutics (prebiotics and probiotics) as well as phages engineered with CRISPR/Cas systems and quorum sensing systems have been developed as new therapies for controlling dysbiosis (alterations in microbial community) and common diseases (e.g., diabetes and obesity). The designing and production of pharmaceuticals based on our own body's microbiome is an emerging field and is rapidly growing to be fully explored in the near future. This review provides an outlook on recent findings on the human microbiomes, their impact on health and diseases, and on the development of targeted therapies.
- 82Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G. A. D.; Gasbarrini, A.; Mele, M. C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14, DOI: 10.3390/microorganisms7010014Google Scholar82https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1ShsbjF&md5=388f6272007c7539cbd95a26581cabefWhat is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseasesRinninella, Emanuele; Raoul, Pauline; Cintoni, Marco; Franceschi, Francesco; Miggiano, Giacinto Abele Donato; Gasbarrini, Antonio; Mele, Maria CristinaMicroorganisms (2019), 7 (1), 14CODEN: MICRKN; ISSN:2076-2607. (MDPI AG)Each individual is provided with a unique gut microbiota profile that plays many specific functions in host nutrient metab., maintenance of structural integrity of the gut mucosal barrier, immunomodulation, and protection against pathogens. Gut microbiota are composed of different bacteria species taxonomically classified by genus, family, order, and phyla. Each human's gut microbiota are shaped in early life as their compn. depends on infant transitions (birth gestational date, type of delivery, methods of milk feeding, weaning period) and external factors such as antibiotic use. These personal and healthy core native microbiota remain relatively stable in adulthood but differ between individuals due to enterotypes, body mass index (BMI) level, exercise frequency, lifestyle, and cultural and dietary habits. Accordingly, there is not a unique optimal gut microbiota compn. since it is different for each individual. However, a healthy host-microorganism balance must be respected in order to optimally perform metabolic and immune functions and prevent disease development. This review will provide an overview of the studies that focus on gut microbiota balances in the same individual and between individuals and highlight the close mutualistic relationship between gut microbiota variations and diseases. Indeed, dysbiosis of gut microbiota is assocd. not only with intestinal disorders but also with numerous extra-intestinal diseases such as metabolic and neurol. disorders. Understanding the cause or consequence of these gut microbiota balances in health and disease and how to maintain or restore a healthy gut microbiota compn. should be useful in developing promising therapeutic interventions.
- 83Cresci, G. A. M.; Izzo, K. Chapter 4 - Gut Microbiome. In Adult Short Bowel Syndrome; Corrigan, M. L., Roberts, K., Steiger, E., Eds.; Academic Press, 2019; pp 45– 54.Google ScholarThere is no corresponding record for this reference.
- 84CAS Content Collection. https://www.cas.org/about/cas-content (accessed January 18, 2023).Google ScholarThere is no corresponding record for this reference.
- 85Kho, Z. Y.; Lal, S. K. The Human Gut Microbiome – A Potential Controller of Wellness and Disease. Front. Microbiol. 2018, 9, 1835, DOI: 10.3389/fmicb.2018.01835Google Scholar85https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3c3islyitA%253D%253D&md5=b3201161709507f19e152a0a0481abf2The Human Gut Microbiome - A Potential Controller of Wellness and DiseaseKho Zhi Y; Lal Sunil KFrontiers in microbiology (2018), 9 (), 1835 ISSN:1664-302X.Interest toward the human microbiome, particularly gut microbiome has flourished in recent decades owing to the rapidly advancing sequence-based screening and humanized gnotobiotic model in interrogating the dynamic operations of commensal microbiota. Although this field is still at a very preliminary stage, whereby the functional properties of the complex gut microbiome remain less understood, several promising findings have been documented and exhibit great potential toward revolutionizing disease etiology and medical treatments. In this review, the interactions between gut microbiota and the host have been focused on, to provide an overview of the role of gut microbiota and their unique metabolites in conferring host protection against invading pathogen, regulation of diverse host physiological functions including metabolism, development and homeostasis of immunity and the nervous system. We elaborate on how gut microbial imbalance (dysbiosis) may lead to dysfunction of host machineries, thereby contributing to pathogenesis and/or progression toward a broad spectrum of diseases. Some of the most notable diseases namely Clostridium difficile infection (infectious disease), inflammatory bowel disease (intestinal immune-mediated disease), celiac disease (multisystemic autoimmune disorder), obesity (metabolic disease), colorectal cancer, and autism spectrum disorder (neuropsychiatric disorder) have been discussed and delineated along with recent findings. Novel therapies derived from microbiome studies such as fecal microbiota transplantation, probiotic and prebiotics to target associated diseases have been reviewed to introduce the idea of how certain disease symptoms can be ameliorated through dysbiosis correction, thus revealing a new scientific approach toward disease treatment. Toward the end of this review, several research gaps and limitations have been described along with suggested future studies to overcome the current research lacunae. Despite the ongoing debate on whether gut microbiome plays a role in the above-mentioned diseases, we have in this review, gathered evidence showing a potentially far more complex link beyond the unidirectional cause-and-effect relationship between them.
- 86Sankar, S. A.; Lagier, J. C.; Pontarotti, P.; Raoult, D.; Fournier, P. E. The human gut microbiome, a taxonomic conundrum. Syst. Appl. Microbiol 2015, 38, 276– 286, DOI: 10.1016/j.syapm.2015.03.004Google Scholar86https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXls1GktL0%253D&md5=c6f708049d43e2ba4bb0111faae14855The human gut microbiome, a taxonomic conundrumSankar, Senthil Alias; Lagier, Jean-Christophe; Pontarotti, Pierre; Raoult, Didier; Fournier, Pierre-EdouardSystematic and Applied Microbiology (2015), 38 (4), 276-286CODEN: SAMIDF; ISSN:0723-2020. (Elsevier GmbH)From culture to metagenomics, within only 130 years, our knowledge of the human microbiome has considerably improved. With >1000 microbial species identified to date, the gastro-intestinal microbiota is the most complex of human biotas. It is composed of a majority of Bacteroidetes and Firmicutes and, although exhibiting great inter-individual variations according to age, geog. origin, disease or antibiotic uptake, it is stable over time. Metagenomic studies have suggested assocns. between specific gut microbiota compns. and a variety of diseases, including irritable bowel syndrome, Crohn's disease, colon cancer, type 2 diabetes and obesity. However, these data remain method-dependent, as no consensus strategy has been defined to decipher the complexity of the gut microbiota. High-throughput culture-independent techniques have highlighted the limitations of culture by showing the importance of uncultured species, whereas modern culture methods have demonstrated that metagenomics underestimates the microbial diversity by ignoring minor populations. In this review, we highlight the progress and challenges that pave the way to a complete understanding of the human gastrointestinal microbiota and its influence on human health.
- 87Manson, J. M.; Rauch, M.; Gilmore, M. S. The Commensal Microbiology of the Gastrointestinal Tract. In GI Microbiota and Regulation of the Immune System; Huffnagle, G. B., Noverr, M. C., Eds.; Landes Bioscience and Springer Science+Business Media, LLC: New York, 2008; pp 15– 28.Google ScholarThere is no corresponding record for this reference.
- 88Kim, D.-H. Gut Microbiota-Mediated Drug-Antibiotic Interactions. Drug Metab. Dispos. 2015, 43, 1581– 1589, DOI: 10.1124/dmd.115.063867Google Scholar88https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhs1Sgu7fF&md5=d479e92f2c129a8ded40ea9b9c743555Gut microbiota-mediated drug-antibiotic interactionsKim, Dong-HyunDrug Metabolism & Disposition (2015), 43 (10), 1581-1589CODEN: DMDSAI; ISSN:1521-009X. (American Society for Pharmacology and Experimental Therapeutics)Xenobiotic metab. involves the biochem. modification of drugs and phytochems. in living organisms, including humans and other animals. In the intestine, the gut microbiota catalyzes the conversion of hydrophilic drugs into absorbable, hydrophobic compds. through hydroxyzation and redn. Drugs and phytochems. are transformed into bioactive (sulfasalazine, lovastatin, and ginsenoside Rb1), bioinactive (chloramphenicol, ranitidine, and metronidazole), and toxic metabolites (nitrazepam), thus affecting the pharmacokinetics of the original compds. Antibiotics suppress the activities of drug-metabolizing enzymes by inhibiting the proliferation of gut microbiota. Antibiotic treatment might influence xenobiotic metabs. more extensively and potently than previously recognized and reduce gut microbiotamediated transformation of orally administered drugs, thereby altering the systemic concns. of intact drugs, their metabolites, or both. This review describes the effects of antibiotics on the metab. of drugs and phytochems. by the gut microbiota.
- 89Deng, P.; Swanson, K. S. Gut microbiota of humans, dogs and cats: current knowledge and future opportunities and challenges. Br. J. Nutr. 2015, 113 (Suppl), S6– 17, DOI: 10.1017/S0007114514002943Google Scholar89https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXjtlSlu7w%253D&md5=3d61cf86b3eea97f65aacc02f12e8306Gut microbiota of humans, dogs and cats: current knowledge and future opportunities and challengesDeng, Ping; Swanson, Kelly S.British Journal of Nutrition (2015), 113 (S1), S6-S17CODEN: BJNUAV; ISSN:0007-1145. (Cambridge University Press)A review. High-throughput DNA sequencing techniques allow for the identification and characterization of microbes and their genes (microbiome). Using these new techniques, microbial populations in several niches of the human body, including the oral and nasal cavities, skin, urogenital tract and gastrointestinal tract, have been described recently. Very little data on the microbiome of companion animals exist, and most of the data have been derived from the anal. of the faeces of healthy lab. animals. High-throughput assays provide opportunities to study the complex and dense populations of the gut microbiota, including bacteria, archaea, fungi, protozoa and viruses. Our lab. and others have recently described the predominant microbial taxa and genes of healthy dogs and cats and how these respond to dietary interventions. In general, faecal microbial phylogeny (e.g. predominance of Firmicutes, Bacteroidetes, Proteobacteria and Actinobacteria) and functional capacity (e.g. major functional groups related to carbohydrate, protein, DNA and vitamin metab.; virulence factors; and cell wall and capsule) of the canine and feline gut are similar to those of the human gut. Initial sequencing projects have provided a glimpse of the microbial super-organism that exists within the canine and feline gut, but leaves much to be explored and discovered. As DNA provides information only about potential functions, studies that focus on the microbial transcriptome, metabolite profiles, and how microbiome changes affect host physiol. and health are clearly required. Future studies must det. how diet compn., antibiotics and other drug therapies, breed and disease affect or are affected by the gut microbiome and how this information may be used to improve diets, identify disease biomarkers and develop targeted disease therapies.
- 90Nardone, G.; Compare, D. The human gastric microbiota: Is it time to rethink the pathogenesis of stomach diseases?. United European Gastroenterol J. 2015, 3, 255– 260, DOI: 10.1177/2050640614566846Google Scholar90https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlyitr3O&md5=a6730cf0d3575ef11cdbeb4572753272The human gastric microbiota: Is it time to rethink the pathogenesis of stomach diseases?Nardone, Gerardo; Compare, DeboraUnited European Gastroenterology Journal (2015), 3 (3), 255-260CODEN: UEGJAZ; ISSN:2050-6414. (Sage Publications Ltd.)Introduction: Although long thought to be a sterile organ, due to its acid prodn., the human stomach holds a core microbiome. Aim: To provide an update of findings related to gastric microbiota and its link with gastric diseases. Methods: We conducted a systematic review of the literature. Results: The development of culture-independent methods facilitated the identification of many bacteria. Five major phyla have been detected in the stomach: Firmicutes, Bacteroidites, Actinobacteria, Fusobacteria and Proteobacteria. At the genera level, the healthy human stomach is dominated by Prevotella, Streptococcus, Veillonella, Rothia and Haemophilus; however, the compn. of the gastric microbiota is dynamic and affected by such factors as diet, drugs and diseases. The interaction between the pre-existing gastric microbiota and Helicobacter pylori infection might influence an individual's risk of gastric disease, including gastric cancer. Conclusions: The maintenance of bacterial homeostasis could be essential for the stomach's health and highlights the chance for therapeutic interventions targeting the gastric microbiota, even if gastric pH, peristalsis and the mucus layer may prevent bacteria colonization; and the definition of gastric microbiota of the healthy stomach is still an ongoing challenging task.
- 91Ghosh, S.; Pramanik, S. Structural diversity, functional aspects and future therapeutic applications of human gut microbiome. Arch. Microbiol. 2021, 203, 5281– 5308, DOI: 10.1007/s00203-021-02516-yGoogle Scholar91https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVaju77K&md5=bf4e1881a45d2b81d40bc769418a59aaStructural diversity, functional aspects and future therapeutic applications of human gut microbiomeGhosh, Soma; Pramanik, SreemantaArchives of Microbiology (2021), 203 (9), 5281-5308CODEN: AMICCW; ISSN:0302-8933. (Springer)Abstr.: The research on human gut microbiome, regarded as the black box of the human body, is still at the stage of infancy as the functional properties of the complex gut microbiome have not yet been understood. Ongoing metagenomic studies have deciphered that the predominant microbial communities belong to eubacterial phyla Firmicutes, Bacteroidetes, Proteobacteria, Fusobacteria, Cyanobacteria, Verrucomicrobia and archaebacterial phylum Euryarchaeota. The indigenous commensal microbial flora prevents opportunistic pathogenic infection and play undeniable roles in digestion, metabolite and signaling mol. prodn. and controlling host's cellular health, immunity and neuropsychiatric behavior. Besides maintaining intestinal health via short-chain fatty acid (SCFA) prodn., gut microbes also aid in neuro-immuno-endocrine modulatory mol. prodn., immune cell differentiation and glucose and lipid metab. Interdependence of diet and intestinal microbial diversity suggests the effectiveness of pre- and pro-biotics in maintenance of gut and systemic health. Several companies worldwide have started potentially exploiting the microbial contribution to human health and have translated their use in disease management and therapeutic applications. The present review discusses the vast diversity of microorganisms playing intricate roles in human metab. The contribution of the intestinal microbiota to regulate systemic activities including gut-brain-immunity crosstalk has been focused. To the best of our knowledge, this review is the first of its kind to collate and discuss the companies worldwide translating the multi-therapeutic potential of human intestinal microbiota, based on the multi-omics studies, i.e. metagenomics and metabolomics, as ready solns. for several metabolic and systemic disorders. Graphic abstr.: [graphic not available: see fulltext].
- 92Leser, T. D.; Mo̷lbak, L. Better living through microbial action: the benefits of the mammalian gastrointestinal microbiota on the host. Environ. Microbiol. 2009, 11, 2194– 2206, DOI: 10.1111/j.1462-2920.2009.01941.xGoogle Scholar92https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1GntrbP&md5=27a4195b0faaab89256c21d3b1bc4e33Better living through microbial action: the benefits of the mammalian gastrointestinal microbiota on the hostLeser, Thomas D.; Moelbak, LarsEnvironmental Microbiology (2009), 11 (9), 2194-2206CODEN: ENMIFM; ISSN:1462-2912. (Wiley-Blackwell)A review. Mammals live in a homeostatic symbiosis with their gastrointestinal microbiota. The mammalian host provides the microbiota with nutrients and a stable environment; whereas the microbiota helps shaping the host's gut mucosa and provides nutritional contributions. Microorganisms start colonizing the gut immediately after birth followed by a succession of populations until a stable, adult microbiota has been established. However, physiol. conditions differ substantially among locations in the gut and det. bacterial d. and diversity. While Firmicutes and Bacteroidetes dominate the gut microbiota in all mammals, the bacterial genera and species diversity is huge and reflects mammalian phylogeny. The main function of the gastrointestinal epithelium is to absorb nutrients and to retain water and electrolytes, yet at the same time it is an efficient barrier against harmful compds. and microorganisms, and is able to neutralize antagonists coincidentally breaching the barrier. These processes are influenced by the microbiota, which modify epithelial expression of genes involved in nutrient uptake and metab., mucosal barrier function, xenobiotic metab., enteric nervous system and motility, hormonal and maturational responses, angiogenesis, cytoskeleton and extracellular matrix, signal transduction, and general cellular functions. Whereas such effects are local at the gut epithelium they may eventually have systemic consequences, e.g. on body wt. and compn.
- 93Linares, D. M.; Ross, P.; Stanton, C. Beneficial Microbes: The pharmacy in the gut. Bioengineered 2016, 7, 11– 20, DOI: 10.1080/21655979.2015.1126015Google Scholar93https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XkvFOjsb0%253D&md5=7070a47d0046a1853be4e8dceedf69aeBeneficial Microbes: The pharmacy in the gutLinares, Daniel M.; Ross, Paul; Stanton, CatherineBioengineered (2016), 7 (1), 11-20CODEN: BIOEGL; ISSN:2165-5987. (Taylor & Francis, Inc.)The scientific evidence supporting the gut microbiome in relation to health maintenance and links with various disease states afflicting humans, from metabolic to mental health, has grown dramatically in the last few years. Strategies addressing the pos. modulation of microbiome functionality assocd. with these disorders offer huge potential to the food and pharmaceutical industries to innovate and provide therapeutic solns. to many of the health issues affecting modern society. Such strategies may involve the use of probiotics and prebiotics as nutritional adjunct therapies. Probiotics are generally recognized to be a good form of therapy to keep harmful, intestinal microorganisms in check, aid digestion and nutrient absorption, and contribute to immune function. Probiotics are reported to improve microbial balance in the intestinal tract and promote the return to a baseline microbial community following a perturbing event (dysbiosis) such as antibiotic therapy. Prebiotics are selectively fermented ingredients that allow specific changes, both in the compn. and/or activity in the gastrointestinal microflora, which confers benefits upon host well-being and health.
- 94Shah, H. N.; Olsen, I.; Bernard, K.; Finegold, S. M.; Gharbia, S.; Gupta, R. S. Approaches to the study of the systematics of anaerobic, Gram-negative, non-sporeforming rods: Current status and perspectives. Anaerobe 2009, 15, 179– 194, DOI: 10.1016/j.anaerobe.2009.08.003Google Scholar94https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlChurfL&md5=7db4a2390acafa75bfbdaab154fa032fApproaches to the study of the systematics of anaerobic, Gram-negative, non-spore forming rods: Current status and perspectivesShah, Haroun N.; Olsen, Ingar; Bernard, Kathy; Finegold, Sydney M.; Gharbia, Saheer; Gupta, Radhey S.Anaerobe (2009), 15 (5), 179-194CODEN: ANAEF8; ISSN:1075-9964. (Elsevier Ltd.)A review. The present article gives an overview of recent taxonomic changes among the Gram-neg., anaerobic rods, briefly highlighting areas where the biol. and ecol. have a bearing on recent nomenclatorial changes. The focus is among the genera Bacteroides, Prevotella, Porphyromonas, Leptotrichia, Dysgonomonas, Fusobacterium and the Synergistes group and addnl. demonstrates the value of conserved indels and group-specific proteins for identifying and circumscribing many of these taxa and the Bacteroidetes-Chlorobi species in general.
- 95Thomas, F.; Hehemann, J.-H.; Rebuffet, E.; Czjzek, M.; Michel, G. Environmental and Gut Bacteroidetes: The Food Connection. Front. Microbiol. 2011, 2, 93, DOI: 10.3389/fmicb.2011.00093Google Scholar95https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3MnntlGnug%253D%253D&md5=0261db88b24d98213748c16711290e03Environmental and gut bacteroidetes: the food connectionThomas Francois; Hehemann Jan-Hendrik; Rebuffet Etienne; Czjzek Mirjam; Michel GurvanFrontiers in microbiology (2011), 2 (), 93 ISSN:.Members of the diverse bacterial phylum Bacteroidetes have colonized virtually all types of habitats on Earth. They are among the major members of the microbiota of animals, especially in the gastrointestinal tract, can act as pathogens and are frequently found in soils, oceans and freshwater. In these contrasting ecological niches, Bacteroidetes are increasingly regarded as specialists for the degradation of high molecular weight organic matter, i.e., proteins and carbohydrates. This review presents the current knowledge on the role and mechanisms of polysaccharide degradation by Bacteroidetes in their respective habitats. The recent sequencing of Bacteroidetes genomes confirms the presence of numerous carbohydrate-active enzymes covering a large spectrum of substrates from plant, algal, and animal origin. Comparative genomics reveal specific Polysaccharide Utilization Loci shared between distantly related members of the phylum, either in environmental or gut-associated species. Moreover, Bacteroidetes genomes appear to be highly plastic and frequently reorganized through genetic rearrangements, gene duplications and lateral gene transfers (LGT), a feature that could have driven their adaptation to distinct ecological niches. Evidence is accumulating that the nature of the diet shapes the composition of the intestinal microbiota. We address the potential links between gut and environmental bacteria through food consumption. LGT can provide gut bacteria with original sets of utensils to degrade otherwise refractory substrates found in the diet. A more complete understanding of the genetic gateways between food-associated environmental species and intestinal microbial communities sheds new light on the origin and evolution of Bacteroidetes as animals' symbionts. It also raises the question as to how the consumption of increasingly hygienic and processed food deprives our microbiota from useful environmental genes and possibly affects our health.
- 96McKee, L. S.; La Rosa, S. L.; Westereng, B.; Eijsink, V. G.; Pope, P. B.; Larsbrink, J. Polysaccharide degradation by the Bacteroidetes: mechanisms and nomenclature. Environ. Microbiol. Rep. 2021, 13, 559– 581, DOI: 10.1111/1758-2229.12980Google Scholar96https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslylt7fK&md5=62fc8423173fefa31728f517b684318cPolysaccharide degradation by the Bacteroidetes: mechanisms and nomenclatureMcKee, Lauren S.; La Rosa, Sabina Leanti; Westereng, Bjoerge; Eijsink, Vincent G.; Pope, Phillip B.; Larsbrink, JohanEnvironmental Microbiology Reports (2021), 13 (5), 559-581CODEN: EMRNAG; ISSN:1758-2229. (Wiley-Blackwell)The Bacteroidetes phylum is renowned for its ability to degrade a wide range of complex carbohydrates, a trait that has enabled its dominance in many diverse environments. The best studied species inhabit the human gut microbiome and use polysaccharide utilization loci (PULs), discrete genetic structures that encode proteins involved in the sensing, binding, deconstruction, and import of target glycans. In many environmental species, polysaccharide degrdn. is tightly coupled to the phylum-exclusive type IX secretion system (T9SS), which is used for the secretion of certain enzymes and is linked to gliding motility. In addn., within specific species these two adaptive systems (PULs and T9SS) are intertwined, with PUL-encoded enzymes being secreted by the T9SS. Here, we discuss the most noteworthy PUL and non-PUL mechanisms that confer specific and rapid polysaccharide degrdn. capabilities to the Bacteroidetes in a range of environments. We also acknowledge that the literature showcasing examples of PULs is rapidly expanding and developing a set of assumptions that can be hard to track back to original findings. Therefore, we present a simple universal description of conserved PUL functions and how they are detd., while proposing a common nomenclature describing PULs and their components, to simplify discussion and understanding of PUL systems.
- 97Ludwig, W.; Schleifer, K.-H.; Whitman, W. B. Revised road map to the phylum Firmicutes. In Bergey’s Manual of Systematic Bacteriology: Vol. Three The Firmicutes; De Vos, P., Garrity, G. M., Jones, D., Krieg, N. R., Ludwig, W., Rainey, F. A., Schleifer, K.-H., Whitman, W. B., Eds.; Springer New York: New York, 2009; pp 1– 13.Google ScholarThere is no corresponding record for this reference.
- 98Yang, M.; Gu, Y.; Li, L.; Liu, T.; Song, X.; Sun, Y.; Cao, X.; Wang, B.; Jiang, K.; Cao, H. Bile Acid–Gut Microbiota Axis in Inflammatory Bowel Disease: From Bench to Bedside. Nutrients 2021, 13, 3143, DOI: 10.3390/nu13093143Google Scholar98https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVKgtrfN&md5=05bbf4ed3aea2831c978ef4758623890Bile Acid-Gut Microbiota Axis in Inflammatory Bowel Disease: From Bench to BedsideYang, Min; Gu, Yu; Li, Lingfeng; Liu, Tianyu; Song, Xueli; Sun, Yue; Cao, Xiaocang; Wang, Bangmao; Jiang, Kui; Cao, HailongNutrients (2021), 13 (9), 3143CODEN: NUTRHU; ISSN:2072-6643. (MDPI AG)A review. Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder of the gastrointestinal tract, with increasing prevalence, and its pathogenesis remains unclear. Accumulating evidence suggested that gut microbiota and bile acids play pivotal roles in intestinal homeostasis and inflammation. Patients with IBD exhibit decreased microbial diversity and abnormal microbial compn. marked by the depletion of phylum Firmicutes (including bacteria involved in bile acid metab.) and the enrichment of phylum Proteobacteria. Dysbiosis leads to blocked bile acid transformation. Thus, the concn. of primary and conjugated bile acids is elevated at the expense of secondary bile acids in IBD. In turn, bile acids could modulate the microbial community. Gut dysbiosis and disturbed bile acids impair the gut barrier and immunity. Several therapies, such as diets, probiotics, prebiotics, engineered bacteria, fecal microbiota transplantation and ursodeoxycholic acid, may alleviate IBD by restoring gut microbiota and bile acids. Thus, the bile acid-gut microbiota axis is closely connected with IBD pathogenesis. Regulation of this axis may be a novel option for treating IBD.
- 99Barka, E. A.; Vatsa, P.; Sanchez, L.; Gaveau-Vaillant, N.; Jacquard, C.; Klenk, H.-P.; Clément, C.; Ouhdouch, Y.; Wezel, G. P. v. Taxonomy, Physiology, and Natural Products of Actinobacteria. Microbiology and Molecular Biology Reviews 2016, 80, 1– 43, DOI: 10.1128/MMBR.00019-15Google Scholar99https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28vls1Sjtg%253D%253D&md5=dac99309cc12275c1dbbc27e457fd9ffTaxonomy, Physiology, and Natural Products of ActinobacteriaBarka Essaid Ait; Vatsa Parul; Sanchez Lisa; Gaveau-Vaillant Nathalie; Jacquard Cedric; Clement Christophe; Meier-Kolthoff Jan P; Klenk Hans-Peter; Ouhdouch Yder; van Wezel Gilles PMicrobiology and molecular biology reviews : MMBR (2016), 80 (1), 1-43 ISSN:.Actinobacteria are Gram-positive bacteria with high G+C DNA content that constitute one of the largest bacterial phyla, and they are ubiquitously distributed in both aquatic and terrestrial ecosystems. Many Actinobacteria have a mycelial lifestyle and undergo complex morphological differentiation. They also have an extensive secondary metabolism and produce about two-thirds of all naturally derived antibiotics in current clinical use, as well as many anticancer, anthelmintic, and antifungal compounds. Consequently, these bacteria are of major importance for biotechnology, medicine, and agriculture. Actinobacteria play diverse roles in their associations with various higher organisms, since their members have adopted different lifestyles, and the phylum includes pathogens (notably, species of Corynebacterium, Mycobacterium, Nocardia, Propionibacterium, and Tropheryma), soil inhabitants (e.g., Micromonospora and Streptomyces species), plant commensals (e.g., Frankia spp.), and gastrointestinal commensals (Bifidobacterium spp.). Actinobacteria also play an important role as symbionts and as pathogens in plant-associated microbial communities. This review presents an update on the biology of this important bacterial phylum.
- 100Hidalgo-Cantabrana, C.; Delgado, S.; Ruiz, L.; Ruas-Madiedo, P.; Sánchez, B.; Margolles, A. Bifidobacteria and Their Health-Promoting Effects. Microbiol Spectr 2017, 5, 5.3.21, DOI: 10.1128/microbiolspec.BAD-0010-2016Google ScholarThere is no corresponding record for this reference.
- 101Stackebrandt, E.; Murray, R. G. E.; Trüper, H. G. Proteobacteria classis nov., a Name for the Phylogenetic Taxon That Includes the “Purple Bacteria and Their Relatives. International Journal of Systematic and Evolutionary Microbiology 1988, 38, 321– 325, DOI: 10.1099/00207713-38-3-321Google ScholarThere is no corresponding record for this reference.
- 102Rizzatti, G.; Lopetuso, L. R.; Gibiino, G.; Binda, C.; Gasbarrini, A. Proteobacteria: A Common Factor in Human Diseases. BioMed. Research International 2017, 2017, 9351507, DOI: 10.1155/2017/9351507Google Scholar102https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MzgsVKgsg%253D%253D&md5=ff061232dd3e1d205818520859b76722Proteobacteria: A Common Factor in Human DiseasesRizzatti G; Lopetuso L R; Gibiino G; Binda C; Gasbarrini ABioMed research international (2017), 2017 (), 9351507 ISSN:.Microbiota represents the entire microbial community present in the gut host. It serves several functions establishing a mutualistic relation with the host. Latest years have seen a burst in the number of studies focusing on this topic, in particular on intestinal diseases. In this scenario, Proteobacteria are one of the most abundant phyla, comprising several known human pathogens. This review highlights the latest findings on the role of Proteobacteria not only in intestinal but also in extraintestinal diseases. Indeed, an increasing amount of data identifies Proteobacteria as a possible microbial signature of disease. Several studies demonstrate an increased abundance of members belonging to this phylum in such conditions. Major evidences currently involve metabolic disorders and inflammatory bowel disease. However, more recent studies suggest a role also in lung diseases, such as asthma and chronic obstructive pulmonary disease, but evidences are still scant. Notably, all these conditions are sustained by various degree of inflammation, which thus represents a core aspect of Proteobacteria-related diseases.
- 103Lee, K.-C.; Webb, R. I.; Janssen, P. H.; Sangwan, P.; Romeo, T.; Staley, J. T.; Fuerst, J. A. Phylum Verrucomicrobia representatives share a compartmentalized cell plan with members of bacterial phylum Planctomycetes. BMC Microbiol. 2009, 9, 5, DOI: 10.1186/1471-2180-9-5Google Scholar103https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1M7msFyitQ%253D%253D&md5=d528d371ef74f6683ff411a93595b264Phylum Verrucomicrobia representatives share a compartmentalized cell plan with members of bacterial phylum PlanctomycetesLee Kuo-Chang; Webb Richard I; Janssen Peter H; Sangwan Parveen; Romeo Tony; Staley James T; Fuerst John ABMC microbiology (2009), 9 (), 5 ISSN:.BACKGROUND: The phylum Verrucomicrobia is a divergent phylum within domain Bacteria including members of the microbial communities of soil and fresh and marine waters; recently extremely acidophilic members from hot springs have been found to oxidize methane. At least one genus, Prosthecobacter, includes species with genes homologous to those encoding eukaryotic tubulins. A significant superphylum relationship of Verrucomicrobia with members of phylum Planctomycetes possessing a unique compartmentalized cell plan, and members of the phylum Chlamydiae including human pathogens with a complex intracellular life cycle, has been proposed. Based on the postulated superphylum relationship, we hypothesized that members of the two separate phyla Planctomycetes and Verrucomicrobia might share a similar ultrastructure plan differing from classical prokaryote organization. RESULTS: The ultrastructure of cells of four members of phylum Verrucomicrobia - Verrucomicrobium spinosum, Prosthecobacter dejongeii, Chthoniobacter flavus, and strain Ellin514 - was examined using electron microscopy incorporating high-pressure freezing and cryosubstitution. These four members of phylum Verrucomicrobia, representing 3 class-level subdivisions within the phylum, were found to possess a compartmentalized cell plan analogous to that found in phylum Planctomycetes. Like all planctomycetes investigated, they possess a major pirellulosome compartment containing a condensed nucleoid and ribosomes surrounded by an intracytoplasmic membrane (ICM), as well as a ribosome-free paryphoplasm compartment between the ICM and cytoplasmic membrane. CONCLUSION: A unique compartmentalized cell plan so far found among Domain Bacteria only within phylum Planctomycetes, and challenging our concept of prokaryote cell plans, has now been found in a second phylum of the Domain Bacteria, in members of phylum Verrucomicrobia. The planctomycete cell plan thus occurs in at least two distinct phyla of the Bacteria, phyla which have been suggested from other evidence to be related phylogenetically in the proposed PVC (Planctomycetes-Verrucomicrobia-Chlamydiae) superphylum. This planctomycete cell plan is present in at least 3 of 6 subdivisions of Verrucomicrobia, suggesting that the common ancestor of the verrucomicrobial phylum was also compartmentalized and possessed such a plan. The presence of this compartmentalized cell plan in both phylum Planctomycetes and phylum Verrucomicrobia suggest that the last common ancestor of these phyla was also compartmentalized.
- 104Rappé, M. S.; Giovannoni, S. J. The Uncultured Microbial Majority. Annu. Rev. Microbiol. 2003, 57, 369– 394, DOI: 10.1146/annurev.micro.57.030502.090759Google Scholar104https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXptFWlsr8%253D&md5=8f5a003db3f5d19683c2ebecfb40e125The uncultured microbial majorityRappe, Michael S.; Giovannoni, Stephen J.Annual Review of Microbiology (2003), 57 (), 369-394CODEN: ARMIAZ; ISSN:0066-4227. (Annual Reviews Inc.)A review. Since the delineation of 12 bacterial phyla by comparative phylogenetic analyses of 16S rRNA in 1987, knowledge of microbial diversity has expanded dramatically owing to the sequencing of rRNA genes cloned from environmental DNA. Currently, only 26 of the approx. 52 identifiable major lineages, or phyla, within the domain Bacteria have cultivated representatives. Evidence from field studies indicates that many of the uncultivated phyla are found in diverse habitats, and some are extraordinarily abundant. In some important environments, including seawater, freshwater, and soil, many biol. and geochem. important organisms are at best only remotely related to any strain that has been characterized by phenotype or by genome sequencing. Genome sequence information that would allow rRNA gene trees to be related to broader patterns in microbial genome evolution is scant, and therefore microbial diversity remains largely unexplored territory.
- 105Geerlings, S. Y.; Ouwerkerk, J. P.; Koehorst, J. J.; Ritari, J.; Aalvink, S.; Stecher, B.; Schaap, P. J.; Paulin, L.; de Vos, W. M.; Belzer, C. Genomic convergence between Akkermansia muciniphila in different mammalian hosts. BMC Microbiol. 2021, 21, 298, DOI: 10.1186/s12866-021-02360-6Google Scholar105https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVeiur%252FJ&md5=fc1edb781f16cb7d3e74f2063be471a8Genomic convergence between Akkermansia muciniphila in different mammalian hostsGeerlings, Sharon Y.; Ouwerkerk, Janneke P.; Koehorst, Jasper J.; Ritari, Jarmo; Aalvink, Steven; Stecher, Baerbel; Schaap, Peter J.; Paulin, Lars; de Vos, Willem M.; Belzer, ClaraBMC Microbiology (2021), 21 (1), 298CODEN: BMMIBC; ISSN:1471-2180. (BioMed Central Ltd.)Akkermansia muciniphila is a member of the human gut microbiota where it resides in the mucus layer and uses mucin as the sole carbon, nitrogen and energy source. A. muciniphila is the only representative of the Verrucomicrobia phylum in the human gut. However, A. muciniphila 16S rRNA gene sequences have also been found in the intestines of many vertebrates. We detected A. muciniphila-like bacteria in the intestines of animals belonging to 15 out of 16 mammalian orders. In addn., other species belonging to the Verrucomicrobia phylum were detected in fecal samples. We isolated 10 new A. muciniphila strains from the feces of chimpanzee, siamang, mouse, pig, reindeer, horse and elephant. The physiol. and genome of these strains were highly similar in comparison to the type strain A. muciniphila MucT. Overall, the genomes of the new strains showed high av. nucleotide identity (93.9 to 99.7%). In these genomes, we detected considerable conservation of at least 75 of the 78 mucin degrdn. genes that were previously detected in the genome of the type strain MucT. The low genomic divergence obsd. in the new strains may indicate that A. muciniphila favors mucosal colonization independent of the differences in hosts. In addn., the conserved mucus degrdn. capability points towards a similar beneficial role of the new strains in regulating host metabolic health.
- 106Liu, M.-J.; Yang, J.-Y.; Yan, Z.-H.; Hu, S.; Li, J.-Q.; Xu, Z.-X.; Jian, Y.-P. Recent findings in Akkermansia muciniphila-regulated metabolism and its role in intestinal diseases. Clin. Nutr. 2022, 41, 2333– 2344, DOI: 10.1016/j.clnu.2022.08.029Google Scholar106https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XisFCqur3N&md5=8427cc10cfe52ddeada931c698ff2bf9Recent findings in Akkermansia muciniphila-regulated metabolism and its role in intestinal diseasesLiu, Meng-Jie; Yang, Jing-Yu; Yan, Zhen-Hua; Hu, Shuang; Li, Jun-Qi; Xu, Zhi-Xiang; Jian, Yong-PingClinical Nutrition (2022), 41 (10), 2333-2344CODEN: CLNUDP; ISSN:0261-5614. (Elsevier Ltd.)A review. The mammalian gastrointestinal tract is colonized with a majority of gut microbes, affecting host metab. and homeostasis. Gut microbiota plays a vital role in nutrient exchange, signaling transduction between intestinal epithelial cells, and resistance to pathogen invasion. Gut microbiota is divided into mucus layer bacteria and intestinal lumen bacteria based on the colonization distribution. Akkermansia muciniphila (A. muciniphila) prefers to colonize in the intestinal mucus layer, and specifically degrades mucins to produce short-chain fatty acids, providing energy for the host and promoting colonization of the bacterium itself. Degrdn. of mucins prompts the host to compensate for the prodn. of more mucins, thereby maintaining the dynamics of these proteins. In the intestinal micro-ecosystem, A. muciniphila is non-pathogenic, and its colonization with suitable abundance contributes to the development of immune system, thus promoting intestinal health. The mechanisms by which A. muciniphila bears a protective role in the host intestine are currently unclear. We summarize the microenvironment for the colonization of A. muciniphila, physiol. characteristics and pathophysiol. impact of A. muciniphila on intestinal diseases, such as irritable bowel syndrome, inflammatory bowel diseases, and intestinal tumors. We also provided updates for current studies on signals that A. muciniphila enhances intestinal barrier integrity and regulates immune response. Together, we conclude that A. muciniphila is a promising probiotic, which could be a microbial target for the treatment of multiple intestinal diseases.
- 107Gupta, R. S.; Sethi, M. Phylogeny and molecular signatures for the phylum Fusobacteria and its distinct subclades. Anaerobe 2014, 28, 182– 198, DOI: 10.1016/j.anaerobe.2014.06.007Google Scholar107https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlCltLvM&md5=e31bb61fa4c7b945081a26c98b3d8573Phylogeny and molecular signatures for the phylum Fusobacteria and its distinct subcladesGupta, Radhey S.; Sethi, MohitAnaerobe (2014), 28 (), 182-198CODEN: ANAEF8; ISSN:1075-9964. (Elsevier Ltd.)The members of the phylum Fusobacteria and its two families, Fusobacteriaceae and Leptotrichiaceae, are distinguished at present mainly on the basis of their branching in the 16S rRNA gene trees and anal. of the internal transcribed spacer sequences in the 16S-23S rDNA. However, no biochem. or mol. characteristics are known that are uniquely shared by all of most members of these groups of bacteria. We report here detailed phylogenetic and comparative analyses on 45 sequenced Fusobacteria genomes to examine their evolutionary relationships and to identify mol. markers that are specific for the members of this phylum. In phylogenetic trees based on 16S rRNA gene sequences or concatenated sequences for 17 conserved proteins, members of the families Fusobacteriaceae and Leptotrichiaceae formed strongly supported clades and were clearly distinguished. In these trees, the species from the genus Fusobacterium also formed a no. of well-supported clades. In parallel, comparative analyses on Fusobacteria genomes have identified 44 conserved signature indels (CSIs) in proteins involved in a broad range of functions that are either specific for the phylum Fusobacteria or a no. of distinct subclades within this phylum. Seven of these CSIs in important proteins are uniquely present in the protein homologs of all sequenced Fusobacteria and they provide potential mol. markers for this phylum. Six and three other CSIs in other protein sequences are specific for members of the families Fusobacteriaceae and Leptotrichiaceae, resp., and they provide novel mol. means for distinguishing members of these two families. Fourteen addnl. CSIs in different proteins, which are specific for either members of the genera Fusobacterium or Leptotrichia, or a no. of other well-supported clades of Fusobacteria at multiple phylogenetic levels, provide mol. markers for these groups and information regarding the evolutionary relationships among the members of this phylum. Lastly, the present work has also identified 14 CSIs in divergent proteins that are specific for three specific subclades of Fusobacterium species, which are also indicated to be distinct by phylogenetic analyses. The members of these three Fusobacterium subclades also differ significantly from each other in their whole genome av. nucleotide identities values, suggesting that they are possible candidates for recognition as different genera. The mol. markers reported here provide novel means for the identification of members of the phylum Fusobacteria and for their classification.
- 108Madhogaria, B.; Bhowmik, P.; Kundu, A. Correlation between human gut microbiome and diseases. Infectious Medicine 2022, 1, 180– 191, DOI: 10.1016/j.imj.2022.08.004Google ScholarThere is no corresponding record for this reference.
- 109Bull, M. J.; Plummer, N. T. Part 1: The Human Gut Microbiome in Health and Disease. Integr. Med. 2014, 13, 17– 22Google Scholar109https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28ngslCqtg%253D%253D&md5=1e800609aae342509788de2879a22170Part 1: The Human Gut Microbiome in Health and DiseaseBull Matthew J; Plummer Nigel TIntegrative medicine (Encinitas, Calif.) (2014), 13 (6), 17-22 ISSN:1546-993X.The bacterial cells harbored within the human gastrointestinal tract (GIT) outnumber the host's cells by a factor of 10 and the genes encoded by the bacteria resident within the GIT outnumber their host's genes by more than 100 times. These human digestive-tract associated microbes are referred to as the gut microbiome. The human gut microbiome and its role in both health and disease has been the subject of extensive research, establishing its involvement in human metabolism, nutrition, physiology, and immune function. Imbalance of the normal gut microbiota have been linked with gastrointestinal conditions such as inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS), and wider systemic manifestations of disease such as obesity, type 2 diabetes, and atopy. In the first part of this review, we evaluate our evolving knowledge of the development, complexity, and functionality of the healthy gut microbiota, and the ways in which the microbial community is perturbed in dysbiotic disease states; the second part of this review covers the role of interventions that have been shown to modulate and stabilize the gut microbiota and also to restore it to its healthy composition from the dysbiotic states seen in IBS, IBD, obesity, type 2 diabetes, and atopy.
- 110Vijay, A.; Valdes, A. M. Role of the gut microbiome in chronic diseases: a narrative review. Eur. J. Clin. Nutr. 2022, 76, 489– 501, DOI: 10.1038/s41430-021-00991-6Google Scholar110https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XjsVOntbc%253D&md5=df621b3f9e2bfd638c58c1209db0de71Role of the gut microbiome in chronic diseases: a narrative reviewVijay, Amrita; Valdes, Ana M.European Journal of Clinical Nutrition (2022), 76 (4), 489-501CODEN: EJCNEQ; ISSN:0954-3007. (Nature Portfolio)A review. The gut microbiome, i.e., the community of bacteria and other microorganisms living in the human gut, has been implicated both directly and indirectly (mediating the effects of diet) on human health. The assocns. between gut microbiome compn. and disease status have been widely reported, while recent studies have demonstrated a role for the gut microbiome in influencing remote organs, mucosal, and immune function. This review details the role of the gut microbiome in chronic diseases and ways it can be modulated for the management or prevention of chronic conditions. The aim of this narrative review is to describe the assocns. between gut microbiome compn. and various types of chronic diseases and to discuss the links to habitual diet and dietary components. Discussed here is the gut microbiome compn. and its assocn. with autoimmune diseases, gut inflammation/bowel disorders, cardiometabolic diseases, chronic kidney disease (CKD). Finally, the gut microbiome is studied as a therapeutic target in the treatment of chronic diseases.
- 111Baquero, F.; Nombela, C. The microbiome as a human organ. Clin Microbiol Infect 2012, 18 (Suppl 4), 2– 4, DOI: 10.1111/j.1469-0691.2012.03916.xGoogle Scholar111https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFOrsr7J&md5=83369e0cc4a735dd21ea26982b23bd9eThe microbiome as a human organBaquero, F.; Nombela, C.Clinical Microbiology and Infection (2012), 18 (Suppl. 4), 2-4CODEN: CMINFM; ISSN:1198-743X. (Wiley-Blackwell)A review. The human organism is a complex structure composed of cells belonging to all three domains of life on Earth, Eukarya, Bacteria and Archaea, as well as their viruses. Bacterial cells of more than a thousand taxonomic units are condensed in a particular functional collective domain, the intestinal microbiome. The microbiome constitutes the last human organ under active research. Like other organs, and despite its intrinsic complexity, the microbiome is readily inherited, in a process probably involving 'small world' power law dynamics of construction in newborns. Like any other organ, the microbiome has physiol. and pathol., and the individual (and collective) health might be damaged when its collective population structure is altered. The diagnostic of microbiomic diseases involves metagenomic studies. The therapeutics of microbiome-induced pathol. include microbiota transplantation, a technique increasingly available. Perhaps a new medical specialty, microbiomol., is being born.
- 112Evans, J. M.; Morris, L. S.; Marchesi, J. R. The gut microbiome: the role of a virtual organ in the endocrinology of the host. J. Endocrinol. 2013, 218, R37– R47, DOI: 10.1530/JOE-13-0131Google Scholar112https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1WltLjF&md5=3ab2a34ea51b8140d352b87e480f1b0aThe gut microbiome: the role of a virtual organ in the endocrinology of the hostEvans, James M.; Morris, Laura S.; Marchesi, Julian R.Journal of Endocrinology (2013), 218 (3), R37-R47CODEN: JOENAK; ISSN:0022-0795. (BioScientifica Ltd.)A review. The human microbiome contains a vast array of microbes and genes that show greater complexity than the host's own karyome; the functions of many of these microbes are beneficial and show co-evolution with the host, while others are detrimental. The microbiota that colonises the gut is now being considered as a virtual organ or emergent system, with properties that need to be integrated into host biol. and physiol. Unlike other organs, the functions that the gut microbiota plays in the host are as yet not fully understood and can be quite easily disrupted by antibiotics, diet or surgery. In this review, we look at some of the best-characterised functions that only the gut microbiota plays and how it interacts with the host's endocrine system and we try to make it clear that the 21st-century biol. cannot afford to ignore this facet of biol., if it wants to fully understand what makes us human.
- 113Haseeb, A.; Shahzad, I.; Ghulam, H.; Muhammad Naeem, F.; Humaira, M.; Imtiaz, M.; Imran, M.; Saima, M.; Muhammad Irfan, U. Gut Microbiome: A New Organ System in Body. In Parasitology and Microbiology Research; Gilberto Antonio Bastidas, P., Asghar Ali, K., Eds.; IntechOpen: Rijeka, Croatia, 2019.Google ScholarThere is no corresponding record for this reference.
- 114Cani, P. D. Human gut microbiome: hopes, threats and promises. Gut 2018, 67, 1716– 1725, DOI: 10.1136/gutjnl-2018-316723Google Scholar114https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisVehtLc%253D&md5=c4f47f74d81d89dee22cc32b466a3f11Human gut microbiome: hopes, threats and promisesCani, Patrice D.Gut (2018), 67 (9), 1716-1725CODEN: GUTTAK; ISSN:0017-5749. (BMJ Publishing Group)A review. The microbiome has received increasing attention over the last 15 years. Although gut microbes have been explored for several decades, investigations of the role of microorganisms that reside in the human gut has attracted much attention beyond classical infectious diseases. For example, numerous studies have reported changes in the gut microbiota during not only obesity, diabetes, and liver diseases but also cancer and even neurodegenerative diseases. The human gut microbiota is viewed as a potential source of novel therapeutics. Between 2013 and 2017, the no. of publications focusing on the gut microbiota was, remarkably, 12 900, which represents four-fifths of the total no. of publications over the last 40 years that investigated this topic. This review discusses recent evidence of the impact of the gut microbiota on metabolic disorders and focus on selected key mechanisms. This review also aims to provide a crit. anal. of the current knowledge in this field, identify putative key issues or problems and discuss misinterpretations. The abundance of metagenomic data generated on comparing diseased and healthy subjects can lead to the erroneous claim that a bacterium is causally linked with the protection or the onset of a disease. In fact, environmental factors such as dietary habits, drug treatments, intestinal motility and stool frequency and consistency are all factors that influence the compn. of the microbiota and should be considered. The cases of the bacteria Prevotella copri and Akkermansia muciniphila will be discussed as key examples.
- 115Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A. N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T. W.; Andrews, E.; Ajami, N. J.; Bonham, K. S.; Brislawn, C. J. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655– 662, DOI: 10.1038/s41586-019-1237-9Google Scholar115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtVOgtL3O&md5=4376c66207c176377e5b973777fba13bMulti-omics of the gut microbial ecosystem in inflammatory bowel diseasesLloyd-Price, Jason; Arze, Cesar; Ananthakrishnan, Ashwin N.; Schirmer, Melanie; Avila-Pacheco, Julian; Poon, Tiffany W.; Andrews, Elizabeth; Ajami, Nadim J.; Bonham, Kevin S.; Brislawn, Colin J.; Casero, David; Courtney, Holly; Gonzalez, Antonio; Graeber, Thomas G.; Hall, A. Brantley; Lake, Kathleen; Landers, Carol J.; Mallick, Himel; Plichta, Damian R.; Prasad, Mahadev; Rahnavard, Gholamali; Sauk, Jenny; Shungin, Dmitry; Vazquez-Baeza, Yoshiki; White, Richard A., III; Braun, Jonathan; Denson, Lee A.; Jansson, Janet K.; Knight, Rob; Kugathasan, Subra; McGovern, Dermot P. B.; Petrosino, Joseph F.; Stappenbeck, Thaddeus S.; Winter, Harland S.; Clish, Clary B.; Franzosa, Eric A.; Vlamakis, Hera; Xavier, Ramnik J.; Huttenhower, CurtisNature (London, United Kingdom) (2019), 569 (7758), 655-662CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Inflammatory bowel diseases, which include Crohn's disease and ulcerative colitis, affect several million individuals worldwide. Crohn's disease and ulcerative colitis are complex diseases that are heterogeneous at the clin., immunol., mol., genetic, and microbial levels. Individual contributing factors have been the focus of extensive research. As part of the Integrative Human Microbiome Project (HMP2 or iHMP), we followed 132 subjects for one year each to generate integrated longitudinal mol. profiles of host and microbial activity during disease (up to 24 time points each; in total 2,965 stool, biopsy, and blood specimens). Here we present the results, which provide a comprehensive view of functional dysbiosis in the gut microbiome during inflammatory bowel disease activity. We demonstrate a characteristic increase in facultative anaerobes at the expense of obligate anaerobes, as well as mol. disruptions in microbial transcription (for example, among clostridia), metabolite pools (acylcarnitines, bile acids, and short-chain fatty acids), and levels of antibodies in host serum. Periods of disease activity were also marked by increases in temporal variability, with characteristic taxonomic, functional, and biochem. shifts. Finally, integrative anal. identified microbial, biochem., and host factors central to this dysregulation. The study's infrastructure resources, results, and data, which are available through the Inflammatory Bowel Disease Multi'omics Database (http://ibdmdb.org), provide the most comprehensive description to date of host and microbial activities in inflammatory bowel diseases.
- 116Mars, R. A. T.; Yang, Y.; Ward, T.; Houtti, M.; Priya, S.; Lekatz, H. R.; Tang, X.; Sun, Z.; Kalari, K. R.; Korem, T. Longitudinal Multi-omics Reveals Subset-Specific Mechanisms Underlying Irritable Bowel Syndrome. Cell 2020, 182, 1460– 1473.e17, DOI: 10.1016/j.cell.2020.08.007Google Scholar116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVajsL3M&md5=75eaf2c7114cfb7251564c30c6687b75Longitudinal multi-omics reveals Subset-Specific Mechanisms Underlying Irritable Bowel SyndromeMars, Ruben A. T.; Yang, Yi; Ward, Tonya; Houtti, Mo; Priya, Sambhawa; Lekatz, Heather R.; Tang, Xiaojia; Sun, Zhifu; Kalari, Krishna R.; Korem, Tal; Bhattarai, Yogesh; Zheng, Tenghao; Bar, Noam; Frost, Gary; Johnson, Abigail J.; van Treuren, Will; Han, Shuo; Ordog, Tamas; Grover, Madhusudan; Sonnenburg, Justin; D'Amato, Mauro; Camilleri, Michael; Elinav, Eran; Segal, Eran; Blekhman, Ran; Farrugia, Gianrico; Swann, Jonathan R.; Knights, Dan; Kashyap, Purna C.Cell (Cambridge, MA, United States) (2020), 182 (6), 1460-1473.e17CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The gut microbiome has been implicated in multiple human chronic gastrointestinal (GI) disorders. Detg. its mechanistic role in disease has been difficult due to apparent disconnects between animal and human studies and lack of an integrated multi-omics view of disease-specific physiol. changes. We integrated longitudinal multi-omics data from the gut microbiome, metabolome, host epigenome, and transcriptome in the context of irritable bowel syndrome (IBS) host physiol. We identified IBS subtype-specific and symptom-related variation in microbial compn. and function. A subset of identified changes in microbial metabolites correspond to host physiol. mechanisms that are relevant to IBS. By integrating multiple data layers, we identified purine metab. as a novel host-microbial metabolic pathway in IBS with translational potential. Our study highlights the importance of longitudinal sampling and integrating complementary multi-omics data to identify functional mechanisms that can serve as therapeutic targets in a comprehensive treatment strategy for chronic GI diseases.
- 117Rinttilä, T.; Lyra, A.; Krogius-Kurikka, L.; Palva, A. Real-time PCR analysis of enteric pathogens from fecal samples of irritable bowel syndrome subjects. Gut Pathog. 2011, 3, 6, DOI: 10.1186/1757-4749-3-6Google Scholar117https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXlsl2ntb0%253D&md5=4f84e5c6102f7ffaea8fb030757f88c3Real-time PCR analysis of enteric pathogens from fecal samples of irritable bowel syndrome subjectsRinttila, Teemu; Lyra, Anna; Krogius-Kurikka, Lotta; Palva, AiriGut Pathogens (2011), 3 (), 6CODEN: GPUAB2; ISSN:1757-4749. (BioMed Central Ltd.)Growing amt. of scientific evidence suggests that microbes are involved in the pathophysiol. of irritable bowel syndrome (IBS). The predominant fecal microbiota compn. of IBS subjects has been widely studied with DNA-based techniques but less research has been focused on the intestinal pathogens in this disorder. Here, we optimized a highly sensitive panel of 12 quant. real-time PCR (qPCR) assays to shed light on the putative presence of intestinal pathogens in IBS sufferers. The panel was used to screen fecal samples from 96 IBS subjects and 23 healthy controls. Fifteen IBS samples (17%) tested pos. for Staphylococcus aureus with a thermonuclease (nuc) gene-targeting qPCR assay, whereas none of the healthy controls were pos. for S. aureus (p < 0.05). The S. aureus-pos. IBS samples were confirmed by sequencing of the PCR amplicons. Clostridium perfringens was detected from IBS and control groups with a similar frequency (13% and 17%, resp.) with α-toxin (plc) gene-targeting qPCR assay while none of the samples tested pos. for the Cl. perfringens enterotoxin-encoding gene (cpe). The qPCR panel consisting of 12 assays for an extensive set of pathogenic microorganisms provides an efficient alternative to the conventional detection of gastrointestinal pathogens and could accelerate the initiation of targeted antibiotic therapy reducing the risk of post-infectious IBS (PI-IBS). S. aureus has not been previously reported to be assocd. with the onset of IBS. Although we discovered significant differences in the prevalence of S. aureus between the study groups, its importance in giving rise to IBS symptoms requires further studies.
- 118Mayer, E. A.; Nance, K.; Chen, S. The Gut-Brain Axis. Annu. Rev. Med. 2022, 73, 439– 453, DOI: 10.1146/annurev-med-042320-014032Google Scholar118https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2cjit1ejsA%253D%253D&md5=e416e85301b2698d90fce325d3075f21The Gut-Brain AxisMayer Emeran A; Nance Karina; Chen ShelleyAnnual review of medicine (2022), 73 (), 439-453 ISSN:.Preclinical evidence has firmly established bidirectional interactions among the brain, the gut, and the gut microbiome. Candidate signaling molecules and at least three communication channels have been identified. Communication within this system is nonlinear, is bidirectional with multiple feedback loops, and likely involves interactions between different channels. Alterations in gut-brain-microbiome interactions have been identified in rodent models of several digestive, psychiatric, and neurological disorders. While alterations in gut-brain interactions have clearly been established in irritable bowel syndrome, a causative role of the microbiome in irritable bowel syndrome remains to be determined. In the absence of specific microbial targets for more effective therapies, current approaches are limited to dietary interventions and centrally targeted pharmacological and behavioral approaches. A more comprehensive understanding of causative influences within the gut-brain-microbiome system and well-designed randomized controlled trials are needed to translate these exciting preclinical findings into effective therapies.
- 119Zhang, T.; Zhang, C.; Zhang, J.; Sun, F.; Duan, L. Efficacy of Probiotics for Irritable Bowel Syndrome: A Systematic Review and Network Meta-Analysis. Front Cell Infect Microbiol 2022, 12, 859967, DOI: 10.3389/fcimb.2022.859967Google Scholar119https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2MvosVOnsQ%253D%253D&md5=435ad6a1bd9f95fbed238df0e858e8d9Efficacy of Probiotics for Irritable Bowel Syndrome: A Systematic Review and Network Meta-AnalysisZhang Tao; Zhang Cunzheng; Zhang Jindong; Duan Liping; Sun Feng; Sun FengFrontiers in cellular and infection microbiology (2022), 12 (), 859967 ISSN:.Background: Irritable bowel syndrome (IBS) is a common gastrointestinal condition. Studies regarding the treatment of IBS with probiotics have not yielded consistent results, and the best probiotics has not yet been confirmed. Therefore, we performed a network meta-analysis (NMA) to assess the relative rank order of different probiotics for IBS. Method: We searched for RCTs on the efficacy of probiotics for IBS until August 25, 2021. The primary outcome was the symptom relief rate, as well as global symptoms, abdominal pain, bloating, and straining scores. The NMA was conducted using Stata 15.0. We also used meta-regression to explore whether the treatment length and dose influenced the efficacy. Results: Forty-three RCTs, with 5,531 IBS patients, were included in this analysis. Firstly, we compared the efficacy of different probiotic species. B.coagulans exhibited the highest probability to be the optimal probiotic specie in improving IBS symptom relief rate, as well as global symptom, abdominal pain, bloating, and straining scores. In regard to the secondary outcomes, L.plantarum ranked first in ameliorating the QOL of IBS patients, but without any significant differences compared with other probiotic species in standardized mean differences (SMD) estimates. Moreover, patients received L.acidophilus had lowest incidence of adverse events. The meta-regression revealed that no significant differences were found between participants using different doses of probiotics in all outcomes, while the treatment length, as a confounder, can significantly influence the efficacy of probiotics in ameliorating abdominal pain (Coef = -2.30; p = 0.035) and straining (Coef = -3.15; p = 0.020) in IBS patients. Thus, we performed the subgroup analysis on treatment length subsequently in these two outcomes, which showed that efficacy of B.coagulans using 8 weeks ranked first both in improving the abdominal pain and straining scores. Additionally, B. coagulans still had significant efficacy compared to different types of probiotic combinations in present study. Conclusions: The findings of this NMA suggested that B.coagulans had prominent efficacy in treating IBS patients, and incorporating B.coagulans into a probiotic combination, or genetically engineering it to amplify its biological function may be a future research target to treat IBS patients. With few direct comparisons available between individual therapies today, this NMA may have utility in forming treatment guideline for IBS with probiotics.
- 120Ponnusamy, K.; Choi, J. N.; Kim, J.; Lee, S. Y.; Lee, C. H. Microbial community and metabolomic comparison of irritable bowel syndrome faeces. J. Med. Microbiol. 2011, 60, 817– 827, DOI: 10.1099/jmm.0.028126-0Google Scholar120https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXotlyktL8%253D&md5=45cb3406def633eeb56142c42ee8112bMicrobial community and metabolomic comparison of irritable bowel syndrome faecesPonnusamy, Kannan; Choi, Jung Nam; Kim, Jiyoung; Lee, Sun-Young; Lee, Choong HwanJournal of Medical Microbiology (2011), 60 (6), 817-827CODEN: JMMIAV; ISSN:0022-2615. (Society for General Microbiology)Human health relies on the compn. of microbiota in an individual's gut and the synthesized metabolites that may alter the gut environment. Gut microbiota and fecal metabolites are involved in several gastrointestinal diseases. In this study, 16S rRNA-specific denaturing gradient gel electrophoresis and quant. PCR anal. showed that the mean similarity of total bacteria was significantly different (P<0.001) in fecal samples from patients with irritable bowel syndrome (IBS; n=11) and from non-IBS (nIBS) patients (n=8). IBS subjects had a significantly higher diversity of total bacteria, as measured by the Shannon index (H') (3.36<H'<4.37, P=0.004), Bacteroidetes and lactobacilli; however, less diversity was obsd. for Bifidobacter (1.7< H'<3.08, P<0.05) and Clostridium coccoides (0.9< H'<2.98, P=0.007). In this study, no significant difference was found in total bacterial quantity (P>0.05). GC/MS-based multi-variate anal. delineated the fecal metabolites of IBS from nIBS samples. Elevated levels of amino acids (alanine and pyroglutamic acid) and phenolic compds. (hydroxyphenyl acetate and hydroxyphenyl propionate) were found in IBS. These results were highly correlated with the abundance of lactobacilli and Clostridium, which indicates an altered metab. rate assocd. with these gut microorganisms. A higher diversity of Bacteroidetes and Lactobacillus groups in IBS fecal samples also correlated with the resp. total quantity. In addn., these changes altered protein and carbohydrate energy metab. in the gut.
- 121Rajilić-Stojanović, M.; Biagi, E.; Heilig, H. G.; Kajander, K.; Kekkonen, R. A.; Tims, S.; de Vos, W. M. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology 2011, 141, 1792– 1801, DOI: 10.1053/j.gastro.2011.07.043Google Scholar121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlOksbrN&md5=da9f508d81243c976bedbd98e3d369ecGlobal and Deep Molecular Analysis of Microbiota Signatures in Fecal Samples From Patients With Irritable Bowel SyndromeRajilic-Stojanovic, Mirjana; Biagi, Elena; Heilig, Hans G. H. J.; Kajander, Kajsa; Kekkonen, Riina A.; Tims, Sebastian; de Vos, Willem M.Gastroenterology (2011), 141 (5), 1792-1801CODEN: GASTAB; ISSN:0016-5085. (Elsevier)Background & Aims: Irritable bowel syndrome (IBS) has been assocd. with disruptions to the intestinal microbiota, but studies have had limited power, coverage, and depth of anal. We aimed to define microbial populations that can be used discriminate the fecal microbiota of patients with IBS from that of healthy subjects and correlate these with IBS intestinal symptom scores. Methods: The microbiota compn. was assessed by global and deep mol. anal. of fecal samples from 62 patients with IBS patients and 46 healthy individuals (controls). We used a comprehensive and highly reproducible phylogenetic microarray in combination with quant. polymerase chain reaction. Results: The intestinal microbiota of IBS patients differed significantly (P = .0005) from that of controls. The microbiota of patients, compared with controls, had a 2-fold increased ratio of the Firmicutes to Bacteroidetes (P = .0002). This resulted from an approx. 1.5-fold increase in nos. of Dorea, Ruminococcus, and Clostridium spp (P < .005); a 2-fold decrease in the no. of Bacteroidetes (P < .0001); a 1.5-fold decrease in nos. of Bifidobacterium and Faecalibacterium spp (P < .05); and, when present, a 4-fold lower av. no. of methanogens (3.50 × 107 vs 8.74 × 106 cells/g feces; P = .003). Correlation anal. of the microbial groups and IBS symptom scores indicated the involvement of several groups of Firmicutes and Proteobacteria in the pathogenesis of IBS. Conclusions: Global and deep mol. anal. of fecal samples indicates that patients with IBS have a different compn. of microbiota. This information might be used to develop better diagnostics and ultimately treatments for IBS.
- 122Pittayanon, R.; Lau, J. T.; Yuan, Y.; Leontiadis, G. I.; Tse, F.; Surette, M.; Moayyedi, P. Gut Microbiota in Patients With Irritable Bowel Syndrome-A Systematic Review. Gastroenterology 2019, 157, 97– 108, DOI: 10.1053/j.gastro.2019.03.049Google Scholar122https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252FhtVaisg%253D%253D&md5=9c44fc6126c5738a4923952192240d91Gut Microbiota in Patients With Irritable Bowel Syndrome-A Systematic ReviewPittayanon Rapat; Lau Jennifer T; Yuan Yuhong; Leontiadis Grigorios I; Tse Frances; Surette Michael; Moayyedi PaulGastroenterology (2019), 157 (1), 97-108 ISSN:.BACKGROUND & AIMS: Irritable bowel syndrome (IBS) is common but difficult to treat. Altering the gut microbiota has been proposed as a strategy for treatment of IBS, but the association between the gut microbiome and IBS symptoms has not been well established. We performed a systematic review to explore evidence for this association. METHODS: We searched databases, including MEDLINE, EMBASE, Cochrane CDSR, and CENTRAL, through April 2, 2018 for case-control studies comparing the fecal or colon microbiomes of adult or pediatric patients with IBS with microbiomes of healthy individuals (controls). The primary outcome was differences in specific gut microbes between patients with IBS and controls. RESULTS: The search identified 2631 citations; 24 studies from 22 articles were included. Most studies evaluated adults presenting with various IBS subtypes. Family Enterobacteriaceae (phylum Proteobacteria), family Lactobacillaceae, and genus Bacteroides were increased in patients with IBS compared with controls, whereas uncultured Clostridiales I, genus Faecalibacterium (including Faecalibacterium prausnitzii), and genus Bifidobacterium were decreased in patients with IBS. The diversity of the microbiota was either decreased or not different in IBS patients compared with controls. More than 40% of included studies did not state whether cases and controls were comparable (did not describe sex and/or age characteristics). CONCLUSIONS: In a systematic review, we identified specific bacteria associated with microbiomes of patients with IBS vs controls. Studies are needed to determine whether these microbes are a product or cause of IBS.
- 123El-Salhy, M.; Hatlebakk, J. G.; Gilja, O. H.; Bråthen Kristoffersen, A.; Hausken, T. Efficacy of faecal microbiota transplantation for patients with irritable bowel syndrome in a randomised, double-blind, placebo-controlled study. Gut 2020, 69, 859– 867, DOI: 10.1136/gutjnl-2019-319630Google Scholar123https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVCht7vI&md5=5ff6c2ba597e8af3db30ac4569f6c021Efficacy of faecal microbiota transplantation for patients with irritable bowel syndrome in a randomised, double-blind, placebo-controlled studyEl-Salhy, Magdy; Hatlebakk, Jan Gunnar; Gilja, Odd Helge; Kristoffersen, Anja Braathen; Hausken, TrygveGut (2020), 69 (5), 859-867CODEN: GUTTAK; ISSN:0017-5749. (BMJ)Objective Faecal microbiota transplantation (FMT) from healthy donors to patients with irritable bowel syndrome (IBS) has been attempted in two previous double-blind, placebo-controlled studies. While one of those studies found improvement of the IBS symptoms, the other found no effect. The present study was conducted to clarify these contradictory findings. Design This randomised, double-blind, placebo-controlled study randomised 165 patients with IBS to placebo (own faeces), 30 g FMT or 60 g FMT at a ratio of 1:1:1. The material for FMT was obtained from one healthy, well-characterised donor, frozen and administered via gastroscope. The primary outcome was a redn. in the IBS symptoms at 3 mo after FMT (response). A response was defined as a decrease of 50 or more points in the total IBS symptom score. The secondary outcome was a redn. in the dysbiosis index (DI) and a change in the intestinal bacterial profile, analyzed by 16S rRNA gene sequencing, at 1 mo following FMT. Results Responses occurred in 23.6%, 76.9% (p < 0.0001) and 89.1% (p < 00.0001) of the patients who received placebo, 30 g FMT and 60 g FMT, resp. These were accompanied by significant improvements in fatigue and the quality of life in patients who received FMT. The intestinal bacterial profiles changed also significantly in the groups received FMT. The FMT adverse events were mild self-limiting gastrointestinal symptoms. Conclusions FMT is an effective treatment for patients with IBS. Utilizing a well-defined donor with a normal DI and favorable specific microbial signature is essential for successful FMT. The response to FMT increases with the dose.
- 124Wilson, B.; Rossi, M.; Kanno, T.; Parkes, G. C.; Anderson, S.; Mason, A. J.; Irving, P. M.; Lomer, M. C.; Whelan, K. β-Galactooligosaccharide in Conjunction With Low FODMAP Diet Improves Irritable Bowel Syndrome Symptoms but Reduces Fecal Bifidobacteria. Am. J. Gastroenterol. 2020, 115, 906– 915, DOI: 10.14309/ajg.0000000000000641Google Scholar124https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38vpsVGgsA%253D%253D&md5=aa2615e105539ac6d3a069d39e76b492β-Galactooligosaccharide in Conjunction With Low FODMAP Diet Improves Irritable Bowel Syndrome Symptoms but Reduces Fecal BifidobacteriaWilson Bridgette; Rossi Megan; Lomer Miranda C; Whelan Kevin; Wilson Bridgette; Lomer Miranda C; Kanno Tokuwa; Mason A James; Parkes Gareth C; Anderson Simon; Irving Peter MThe American journal of gastroenterology (2020), 115 (6), 906-915 ISSN:.INTRODUCTION: The low FODMAP diet (LFD) reduces symptoms and bifidobacteria in irritable bowel syndrome (IBS). β-galactooligosaccharides (B-GOS) may reduce the symptoms and increase bifidobacteria in IBS. We investigated whether B-GOS supplementation alongside the LFD improves IBS symptoms while preventing the decline in bifidobacteria. METHODS: We performed a randomized, placebo-controlled, 3-arm trial of 69 Rome III adult patients with IBS from secondary care in the United Kingdom. Patients were randomized to a sham diet with placebo supplement (control) or LFD supplemented with either placebo (LFD) or 1.4 g/d B-GOS (LFD/B-GOS) for 4 weeks. Gastrointestinal symptoms, fecal microbiota (fluorescent in situ hybridization and 16S rRNA sequencing), fecal short-chain fatty acids (gas-liquid chromatography) and pH (probe), and urine metabolites (H NMR) were analyzed. RESULTS: At 4 weeks, adequate symptom relief was higher in the LFD/B-GOS group (16/24, 67%) than in the control group (7/23, 30%) (odds ratio 4.6, 95% confidence interval: 1.3-15.6; P = 0.015); Bifidobacterium concentrations (log10 cells/g dry weight) were not different between LFD and LFD/B-GOS but were lower in the LFD/B-GOS (9.49 [0.73]) than in the control (9.77 [0.41], P = 0.018). A proportion of Actinobacteria was lower in LFD (1.9%, P = 0.003) and LFD/B-GOS (1.8%, P < 0.001) groups than in the control group (4.2%). Fecal butyrate was lower in the LFD (387.3, P = 0.028) and LFD/B-GOS (346.0, P = 0.007) groups than in the control group (609.2). DISCUSSION: The LFD combined with B-GOS prebiotic produced a greater symptom response than the sham diet plus placebo, but addition of 1.4 g/d B-GOS did not prevent the reduction of bifidobacteria. The LFD reduces fecal Actinobacteria and butyrate thus strict long-term use should not be advised.
- 125Vasant, D. H.; Paine, P. A.; Black, C. J.; Houghton, L. A.; Everitt, H. A.; Corsetti, M.; Agrawal, A.; Aziz, I.; Farmer, A. D.; Eugenicos, M. P. British Society of Gastroenterology guidelines on the management of irritable bowel syndrome. Gut 2021, 70, 1214– 1240, DOI: 10.1136/gutjnl-2021-324598Google Scholar125https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3sblvVektg%253D%253D&md5=3d4f2e18c2008dea28e43666bd713126British Society of Gastroenterology guidelines on the management of irritable bowel syndromeVasant Dipesh H; Vasant Dipesh H; Paine Peter A; Paine Peter A; Black Christopher J; Ford Alexander C; Black Christopher J; Houghton Lesley A; Ford Alexander C; Houghton Lesley A; Everitt Hazel A; Corsetti Maura; Agrawal Anurag; Aziz Imran; Aziz Imran; Farmer Adam D; Farmer Adam D; Eugenicos Maria P; Moss-Morris Rona; Yiannakou YanGut (2021), 70 (7), 1214-1240 ISSN:.Irritable bowel syndrome (IBS) remains one of the most common gastrointestinal disorders seen by clinicians in both primary and secondary care. Since publication of the last British Society of Gastroenterology (BSG) guideline in 2007, substantial advances have been made in understanding its complex pathophysiology, resulting in its re-classification as a disorder of gut-brain interaction, rather than a functional gastrointestinal disorder. Moreover, there has been a considerable amount of new evidence published concerning the diagnosis, investigation and management of IBS. The primary aim of this guideline, commissioned by the BSG, is to review and summarise the current evidence to inform and guide clinical practice, by providing a practical framework for evidence-based management of patients. One of the strengths of this guideline is that the recommendations for treatment are based on evidence derived from a comprehensive search of the medical literature, which was used to inform an update of a series of trial-based and network meta-analyses assessing the efficacy of dietary, pharmacological and psychological therapies in treating IBS. Specific recommendations have been made according to the Grading of Recommendations Assessment, Development and Evaluation system, summarising both the strength of the recommendations and the overall quality of evidence. Finally, this guideline identifies novel treatments that are in development, as well as highlighting areas of unmet need for future research.
- 126Moayyedi, P.; Andrews, C. N.; MacQueen, G.; Korownyk, C.; Marsiglio, M.; Graff, L.; Kvern, B.; Lazarescu, A.; Liu, L.; Paterson, W. G. Canadian Association of Gastroenterology Clinical Practice Guideline for the Management of Irritable Bowel Syndrome (IBS). J. Can. Assoc Gastroenterol 2019, 2, 6– 29, DOI: 10.1093/jcag/gwy071Google Scholar126https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3Mzmt1OnsA%253D%253D&md5=0774e7ab2ec3948ad8c141e21c61867aCanadian Association of Gastroenterology Clinical Practice Guideline for the Management of Irritable Bowel Syndrome (IBS)Moayyedi Paul; Sidani Sacha; Andrews Christopher N; MacQueen Glenda; Korownyk Christina; Marsiglio Megan; Graff Lesley; Kvern Brent; Lazarescu Adriana; Liu Louis; Paterson William G; Vanner StephenJournal of the Canadian Association of Gastroenterology (2019), 2 (1), 6-29 ISSN:.BACKGROUND & AIMS: Irritable bowel syndrome (IBS) is one of the most common gastrointestinal (GI) disorders, affecting about 10% of the general population globally. The aim of this consensus was to develop guidelines for the management of IBS. METHODS: A systematic literature search identified studies on the management of IBS. The quality of evidence and strength of recommendations were rated according to the Grading of Recommendation Assessment, Development and Evaluation (GRADE) approach. Statements were developed through an iterative online platform and then finalized and voted on by a multidisciplinary group of clinicians and a patient. RESULTS: Consensus was reached on 28 of 31 statements. Irritable bowel syndrome is diagnosed based on symptoms; serological testing is suggested to exclude celiac disease, but routine testing for C-reactive protein (CRP), fecal calprotectin or food allergies is not recommended. A trial of a low fermentable oligosaccharides, disaccharides, monosaccharides, polyols (FODMAP) diet is suggested, while a gluten-free diet is not. Psyllium, but not wheat bran, supplementation may help reduce symptoms. Alternative therapies such as peppermint oil and probiotics are suggested, while herbal therapies and acupuncture are not. Cognitive behavioural therapy and hypnotherapy are suggested psychological therapies. Among the suggested or recommended pharmacological therapies are antispasmodics, certain antidepressants, eluxadoline, lubiprostone, and linaclotide. Loperamide, cholestyramine and osmotic laxatives are not recommended for overall IBS symptoms. The nature of the IBS symptoms (diarrhea-predominant or constipation-predominant) should be considered in the choice of pharmacological treatments. CONCLUSIONS: Patients with IBS may benefit from a multipronged, individualized approach to treatment, including dietary modifications, psychological and pharmacological therapies.
- 127Fukudo, S.; Okumura, T.; Inamori, M.; Okuyama, Y.; Kanazawa, M.; Kamiya, T.; Sato, K.; Shiotani, A.; Naito, Y.; Fujikawa, Y. Evidence-based clinical practice guidelines for irritable bowel syndrome 2020. J. Gastroenterol. 2021, 56, 193– 217, DOI: 10.1007/s00535-020-01746-zGoogle Scholar127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3srpsFajtg%253D%253D&md5=1e7734581ce8e2ac14b0c96312d3c01aEvidence-based clinical practice guidelines for irritable bowel syndrome 2020Fukudo Shin; Okumura Toshikatsu; Inamori Masahiko; Okuyama Yusuke; Kanazawa Motoyori; Kamiya Takeshi; Sato Ken; Shiotani Akiko; Naito Yuji; Fujikawa Yoshiko; Hokari Ryota; Masaoka Tastuhiro; Fujimoto Kazuma; Kaneko Hiroshi; Torii Akira; Matsueda Kei; Miwa Hiroto; Enomoto Nobuyuki; Shimosegawa Tooru; Koike Kazuhiko; Fukudo ShinJournal of gastroenterology (2021), 56 (3), 193-217 ISSN:.Managing irritable bowel syndrome (IBS) has attracted international attention because single-agent therapy rarely relieves bothersome symptoms for all patients. The Japanese Society of Gastroenterology (JSGE) published the first edition of evidence-based clinical practice guidelines for IBS in 2015. Much more evidence has accumulated since then, and new pharmacological agents and non-pharmacological methods have been developed. Here, we report the second edition of the JSGE-IBS guidelines comprising 41 questions including 12 background questions on epidemiology, pathophysiology, and diagnostic criteria, 26 clinical questions on diagnosis and treatment, and 3 questions on future research. For each question, statements with or without recommendations and/or evidence level are given and updated diagnostic and therapeutic algorithms are provided based on new evidence. Algorithms for diagnosis are requisite for patients with chronic abdominal pain or associated symptoms and/or abnormal bowel movement. Colonoscopy is indicated for patients with one or more alarm symptoms/signs, risk factors, and/or abnormal routine examination results. The diagnosis is based on the Rome IV criteria. Step 1 therapy consists of diet therapy, behavioral modification, and gut-targeted pharmacotherapy for 4 weeks. For non-responders, management proceeds to step 2 therapy, which includes a combination of different mechanistic gut-targeted agents and/or psychopharmacological agents and basic psychotherapy for 4 weeks. Step 3 therapy is for non-responders to step 2 and comprises a combination of gut-targeted pharmacotherapy, psychopharmacological treatments, and/or specific psychotherapy. These updated JSGE-IBS guidelines present best practice strategies for IBS patients in Japan and we believe these core strategies can be useful for IBS diagnosis and treatment globally.
- 128Lacy, B. E.; Pimentel, M.; Brenner, D. M.; Chey, W. D.; Keefer, L. A.; Long, M. D.; Moshiree, B. ACG Clinical Guideline: Management of Irritable Bowel Syndrome. Am. J. Gastroenterol. 2021, 116, 17, DOI: 10.14309/ajg.0000000000001036Google Scholar128https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXnt1Grtrk%253D&md5=26264e26c18a80d33974d3dbdddf4303ACG clinical guideline: management of irritable bowel syndromeLacy, Brian E.; Pimentel, Mark; Brenner, Darren M.; Chey, William D.; Keefer, Laurie A.; Long, Millie D.; Moshiree, BahaAmerican Journal of Gastroenterology (2021), 116 (1), 17-44CODEN: AJGAAR; ISSN:1572-0241. (Wolters Kluwer)Irritable bowel syndrome (IBS) is a highly prevalent, chronic disorder that significantly reduces patients' quality of life. Advances in diagnostic testing and in therapeutic options for patients with IBS led to the development of this first-ever American College of Gastroenterol. clin. guideline for the management of IBS using Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) methodol. Twenty-five clin. important questions were assessed after a comprehensive literature search; 9 questions focused on diagnostic testing; 16 questions focused on therapeutic options. Consensus was obtained using a modified Delphi approach, and based on GRADE methodol., we endorse the following: We suggest that a pos. diagnostic strategy as compared to a diagnostic strategy of exclusion be used to improve time to initiating appropriate therapy. We suggest that serol. testing be performed to rule out celiac disease in patients with IBS and diarrhea symptoms. We suggest that fecal calprotectin be checked in patients with suspected IBS and diarrhea symptoms to rule out inflammatory bowel disease. We recommend a limited trial of a low fermentable oligosaccharides, disacchardies, monosaccharides, polyols (FODMAP) diet in patients with IBS to improve global symptoms. We recommend the use of chloride channel activators and guanylate cyclase activators to treat global IBS with constipation symptoms. We recommend the use of rifaximin to treat global IBS with diarrhea symptoms. We suggest that gut-directed psychotherapy be used to treat global IBS symptoms. Addnl. statements and information regarding diagnostic strategies, specific drugs, doses, and duration of therapy can be found in the guideline.
- 129Ford, A. C.; Harris, L. A.; Lacy, B. E.; Quigley, E. M. M.; Moayyedi, P. Systematic review with meta-analysis: the efficacy of prebiotics, probiotics, synbiotics and antibiotics in irritable bowel syndrome. Aliment. Pharmacol. Ther. 2018, 48, 1044– 1060, DOI: 10.1111/apt.15001Google Scholar129https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3czmslWmuw%253D%253D&md5=f74e29349f47d2004d956c06846f4691Systematic review with meta-analysis: the efficacy of prebiotics, probiotics, synbiotics and antibiotics in irritable bowel syndromeFord Alexander C; Ford Alexander C; Harris Lucinda A; Lacy Brian E; Quigley Eamonn M M; Moayyedi PaulAlimentary pharmacology & therapeutics (2018), 48 (10), 1044-1060 ISSN:.BACKGROUND: Irritable bowel syndrome (IBS) is a chronic functional bowel disorder. Disturbances in the gastrointestinal microbiome may be involved in its aetiology. AIM: To perform a systematic review and meta-analysis to examine the efficacy of prebiotics, probiotics, synbiotics and antibiotics in IBS. METHODS: MEDLINE, EMBASE, and the Cochrane Controlled Trials Register were searched (up to July 2017). Randomised controlled trials (RCTs) recruiting adults with IBS, comparing prebiotics, probiotics, synbiotics or antibiotics with placebo or no therapy were eligible. Dichotomous symptom data were pooled to obtain a relative risk (RR) of remaining symptomatic after therapy, with a 95% confidence interval (CI). Continuous data were pooled using a standardised mean difference with a 95% CI. RESULTS: The search identified 4017 citations. Data for prebiotics and synbiotics were sparse. Fifty-three RCTs of probiotics, involving 5545 patients, were eligible. Particular combinations of probiotics, or specific species and strains, appeared to have beneficial effects on global IBS symptoms and abdominal pain, but it was not possible to draw definitive conclusions about their efficacy. There were five trials of similar design that used rifaximin in non-constipated IBS patients, which was more effective than placebo (RR of symptoms persisting = 0.84; 95% CI 0.79-0.90). Adverse events were no more common with probiotics or antibiotics. CONCLUSIONS: Which particular combination, species or strains of probiotics are effective for IBS remains, for the most part, unclear. Rifaximin has modest efficacy in improving symptoms in non-constipated IBS.
- 130Caruso, R.; Lo, B. C.; Núñez, G. Host-microbiota interactions in inflammatory bowel disease. Nat. Rev. Immunol. 2020, 20, 411– 426, DOI: 10.1038/s41577-019-0268-7Google Scholar130https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXislequ70%253D&md5=3ebd4c5a2950e065f9c578324d0643b5Host-microbiota interactions in inflammatory bowel diseaseCaruso, Roberta; Lo, Bernard C.; Nunez, GabrielNature Reviews Immunology (2020), 20 (7), 411-426CODEN: NRIABX; ISSN:1474-1733. (Nature Research)Abstr.: The mammalian intestine is colonized by trillions of microorganisms that have co-evolved with the host in a symbiotic relationship. The presence of large nos. of symbionts near the epithelial surface of the intestine poses an enormous challenge to the host because it must avoid the activation of harmful inflammatory responses to the microorganisms while preserving its ability to mount robust immune responses to invading pathogens. In patients with inflammatory bowel disease, there is a breakdown of the multiple strategies that the immune system has evolved to promote the sepn. between symbiotic microorganisms and the intestinal epithelium and the effective killing of penetrant microorganisms, while suppressing the activation of inappropriate T cell responses to resident microorganisms. Understanding the complex interactions between intestinal microorganisms and the host may provide crucial insight into the pathogenesis of inflammatory bowel disease as well as new avenues to prevent and treat the disease.
- 131Federici, S.; Kredo-Russo, S.; Valdés-Mas, R.; Kviatcovsky, D.; Weinstock, E.; Matiuhin, Y.; Silberberg, Y.; Atarashi, K.; Furuichi, M.; Oka, A. Targeted suppression of human IBD-associated gut microbiota commensals by phage consortia for treatment of intestinal inflammation. Cell 2022, 185, 2879– 2898.e24, DOI: 10.1016/j.cell.2022.07.003Google Scholar131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitVChurfF&md5=e0fa16b8aea85b8df9c03a9e7ca246d8Targeted suppression of human IBD-associated gut microbiota commensals by phage consortia for treatment of intestinal inflammationFederici, Sara; Kredo-Russo, Sharon; Valdes-Mas, Rafael; Kviatcovsky, Denise; Weinstock, Eyal; Matiuhin, Yulia; Silberberg, Yael; Atarashi, Koji; Furuichi, Munehiro; Oka, Akihiko; Liu, Bo; Fibelman, Morine; Weiner, Iddo Nadav; Khabra, Efrat; Cullin, Nyssa; Ben-Yishai, Noa; Inbar, Dana; Ben-David, Hava; Nicenboim, Julian; Kowalsman, Noga; Lieb, Wolfgang; Kario, Edith; Cohen, Tal; Geffen, Yael Friedman; Zelcbuch, Lior; Cohen, Ariel; Rappo, Urania; Gahali-Sass, Inbar; Golembo, Myriam; Lev, Vered; Dori-Bachash, Mally; Shapiro, Hagit; Moresi, Claudia; Cuevas-Sierra, Amanda; Mohapatra, Gayatree; Kern, Lara; Zheng, Danping; Nobs, Samuel Philip; Suez, Jotham; Stettner, Noa; Harmelin, Alon; Zak, Naomi; Puttagunta, Sailaja; Bassan, Merav; Honda, Kenya; Sokol, Harry; Bang, Corinna; Franke, Andre; Schramm, Christoph; Maharshak, Nitsan; Sartor, Ryan Balfour; Sorek, Rotem; Elinav, EranCell (Cambridge, MA, United States) (2022), 185 (16), 2879-2898.e24CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Human gut commensals are increasingly suggested to impact non-communicable diseases, such as inflammatory bowel diseases (IBD), yet their targeted suppression remains a daunting unmet challenge. In four geog. distinct IBD cohorts (n = 537), we identify a clade of Klebsiella pneumoniae (Kp) strains, featuring a unique antibiotics resistance and mobilome signature, to be strongly assocd. with disease exacerbation and severity. Transfer of clin. IBD-assocd. Kp strains into colitis-prone, germ-free, and colonized mice enhances intestinal inflammation. Stepwise generation of a lytic five-phage combination, targeting sensitive and resistant IBD-assocd. Kp clade members through distinct mechanisms, enables effective Kp suppression in colitis-prone mice, driving an attenuated inflammation and disease severity. Proof-of-concept assessment of Kp-targeting phages in an artificial human gut and in healthy volunteers demonstrates gastric acid-dependent phage resilience, safety, and viability in the lower gut. Collectively, we demonstrate the feasibility of orally administered combination phage therapy in avoiding resistance, while effectively inhibiting non-communicable disease-contributing pathobionts.
- 132Nagalingam, N. A.; Lynch, S. V. Role of the microbiota in inflammatory bowel diseases. Inflamm. Bowel Dis. 2012, 18, 968– 984, DOI: 10.1002/ibd.21866Google Scholar132https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38zlvFehuw%253D%253D&md5=36eaa38f264de538590085654a704292Role of the microbiota in inflammatory bowel diseasesNagalingam Nabeetha A; Lynch Susan VInflammatory bowel diseases (2012), 18 (5), 968-84 ISSN:.Studying the role of the human microbiome as it relates to human health status has revolutionized our view of microbial community contributions to a large number of diseases, particularly chronic inflammatory disorders. The lower gastrointestinal (GI) tract houses trillions of microbial cells representing a large diversity of species in relatively well-defined phylogenetic ratios that are associated with maintenance of key aspects of host physiology and immune homeostasis. It is not surprising, therefore, that many GI inflammatory diseases, including inflammatory bowel disease (IBD), are associated with substantial changes in the composition of these microbial assemblages, either as a cause or consequence of host inflammatory response. Here we review current knowledge in the emerging field of human microbiome research as it relates to IBD, specifically focusing on Crohn's disease (CD) and ulcerative colitis (UC). We discuss bacteriotherapeutic efforts to restore GI microbial assemblage integrity via probiotic supplementation of IBD patients, and speculate on future directions for the field.
- 133Barcenilla, A.; Pryde, S. E.; Martin, J. C.; Duncan, S. H.; Stewart, C. S.; Henderson, C.; Flint, H. J. Phylogenetic relationships of butyrate-producing bacteria from the human gut. Appl. Environ. Microbiol. 2000, 66, 1654– 1661, DOI: 10.1128/AEM.66.4.1654-1661.2000Google Scholar133https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXisVWmu7g%253D&md5=98a4294d41eb8141fdda17b857791129Phylogenetic relationships of butyrate-producing bacteria from the human gutBarcenilla, Adela; Pryde, Susan E.; Martin, Jennifer C.; Duncan, Sylvia H.; Stewart, Colin S.; Henderson, Colin; Flint, Harry J.Applied and Environmental Microbiology (2000), 66 (4), 1654-1661CODEN: AEMIDF; ISSN:0099-2240. (American Society for Microbiology)Butyrate is a preferred energy source for colonic epithelial cells and is thought to play an important role in maintaining colonic health in humans. In order to investigate the diversity and stability of butyrate-producing organisms of the colonic flora, anaerobic butyrate-producing bacteria were isolated from freshly voided human fecal samples from 3 healthy individuals: an infant, an adult omnivore, and an adult vegetarian. A 2nd isolation was performed on the same 3 individuals 1 yr later. Of a total of 313 bacterial isolates, 74 produced more than 2 mM butyrate in vitro. Butyrate-producing isolates were grouped by 16S ribosomal DNA (rDNA) PCR-restriction fragment length polymorphism anal. The results indicate very little overlap between the predominant ribotypes of the 3 subjects; furthermore, the flora of each individual changed significantly between the 2 isolations. Complete sequences of 16S rDNAs were detd. for 24 representative strains and subjected to phylogenetic anal. Eighty percent of the butyrate-producing isolates fell within the XIVa cluster of gram-pos. bacteria as defined by M. D. Collins et al. and A. Willems et al., with the most abundant group (10 of 24 or 42%) clustering with Eubacterium rectale, Eubacterium ramulus, and Roseburia cecicola. Fifty percent of the butyrate-producing isolates were net acetate consumers during growth, suggesting that they employ the butyryl CoA-acetyl CoA transferase pathway for butyrate prodn. In contrast, only 1% of the 239 non-butyrate-producing isolates consumed acetate.
- 134Halfvarson, J.; Brislawn, C. J.; Lamendella, R.; Vázquez-Baeza, Y.; Walters, W. A.; Bramer, L. M.; D’Amato, M.; Bonfiglio, F.; McDonald, D.; Gonzalez, A. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat. Microbiol 2017, 2, 17004, DOI: 10.1038/nmicrobiol.2017.4Google Scholar134https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXkvFyqt7s%253D&md5=31d20fb740e9e065969614896c2cca3aDynamics of the human gut microbiome in inflammatory bowel diseaseHalfvarson, Jonas; Brislawn, Colin J.; Lamendella, Regina; Vazquez-Baeza, Yoshiki; Walters, William A.; Bramer, Lisa M.; D'Amato, Mauro; Bonfiglio, Ferdinando; McDonald, Daniel; Gonzalez, Antonio; McClure, Erin E.; Dunklebarger, Mitchell F.; Knight, Rob; Jansson, Janet K.Nature Microbiology (2017), 2 (1), 17004CODEN: NMAICH; ISSN:2058-5276. (Nature Publishing Group)Inflammatory bowel disease (IBD) is characterized by flares of inflammation with a periodic need for increased medication and sometimes even surgery. The etiology of IBD is partly attributed to a deregulated immune response to gut microbiome dysbiosis. Cross-sectional studies have revealed microbial signatures for different IBD subtypes, including ulcerative colitis, colonic Crohn's disease and ileal Crohn's disease. Although IBD is dynamic, microbiome studies have primarily focused on single time points or a few individuals. Here, we dissect the long-term dynamic behavior of the gut microbiome in IBD and differentiate this from normal variation. Microbiomes of IBD subjects fluctuate more than those of healthy individuals, based on deviation from a newly defined healthy plane (HP). Ileal Crohn's disease subjects deviated most from the HP, esp. subjects with surgical resection. Intriguingly, the microbiomes of some IBD subjects periodically visited the HP then deviated away from it. Inflammation was not directly correlated with distance to the healthy plane, but there was some correlation between obsd. dramatic fluctuations in the gut microbiome and intensified medication due to a flare of the disease. These results will help guide therapies that will redirect the gut microbiome towards a healthy state and maintain remission in IBD.
- 135Mottawea, W.; Chiang, C.-K.; Mühlbauer, M.; Starr, A. E.; Butcher, J.; Abujamel, T.; Deeke, S. A.; Brandel, A.; Zhou, H.; Shokralla, S. Altered intestinal microbiota–host mitochondria crosstalk in new onset Crohn’s disease. Nat. Commun. 2016, 7, 13419, DOI: 10.1038/ncomms13419Google Scholar135https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFSns77P&md5=700643e7cd70e13bb35625e955a3bce7Altered intestinal microbiota-host mitochondria crosstalk in new onset Crohn's diseaseMottawea, Walid; Chiang, Cheng-Kang; Muhlbauer, Marcus; Starr, Amanda E.; Butcher, James; Abujamel, Turki; Deeke, Shelley A.; Brandel, Annette; Zhou, Hu; Shokralla, Shadi; Hajibabaei, Mehrdad; Singleton, Ruth; Benchimol, Eric I.; Jobin, Christian; Mack, David R.; Figeys, Daniel; Stintzi, AlainNature Communications (2016), 7 (), 13419CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Intestinal microbial dysbiosis is assocd. with Crohn's disease (CD). However, the mechanisms leading to the chronic mucosal inflammation that characterizes this disease remain unclear. In this report, we use systems-level approaches to study the interactions between the gut microbiota and host in new-onset paediatric patients to evaluate causality and mechanisms of disease. We report an altered host proteome in CD patients indicative of impaired mitochondrial functions. In particular, mitochondrial proteins implicated in H2S detoxification are downregulated, while the relative abundance of H2S microbial producers is increased. Network correlation anal. reveals that Atopobium parvulum controls the central hub of H2S producers. A. parvulum induces pancolitis in colitis-susceptible interleukin-10-deficient mice and this phenotype requires the presence of the intestinal microbiota. Administrating the H2S scavenger bismuth mitigates A. parvulum-induced colitis in vivo. This study reveals that host-microbiota interactions are disturbed in CD and thus provides mechanistic insights into CD pathogenesis.
- 136Marchesi, J. R.; Holmes, E.; Khan, F.; Kochhar, S.; Scanlan, P.; Shanahan, F.; Wilson, I. D.; Wang, Y. Rapid and noninvasive metabonomic characterization of inflammatory bowel disease. J. Proteome Res. 2007, 6, 546– 551, DOI: 10.1021/pr060470dGoogle Scholar136https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhs12gtQ%253D%253D&md5=af033580ca436e4056e7262110730ac1Rapid and Noninvasive Metabonomic Characterization of Inflammatory Bowel DiseaseMarchesi, Julian R.; Holmes, Elaine; Khan, Fatima; Kochhar, Sunil; Scanlan, Pauline; Shanahan, Fergus; Wilson, Ian D.; Wang, YulanJournal of Proteome Research (2007), 6 (2), 546-551CODEN: JPROBS; ISSN:1535-3893. (American Chemical Society)Inflammatory bowel diseases (IBD) including Crohn's disease (CD) and ulcerative colitis (UC) have a major impact on the health of individuals and populations. Accurate diagnosis of inflammatory bowel disease (IBD) at an early stage, and correct differentiation between Crohn's disease (CD) and ulcerative colitis (UC), is important for optimum treatment and prognosis. The authors present here the first characterization of fecal exts. obtained from patients with CD and UC by employing a noninvasive metabonomics approach, which combines high resoln. 1H NMR spectroscopy and multivariate pattern recognition techniques. The fecal exts. of both CD and UC patients were characterized by reduced levels of butyrate, acetate, methylamine, and trimethylamine in comparison with a control population, suggesting changes in the gut microbial community. Also, elevated quantities of amino acids were present in the feces from both disease groups, implying malabsorption caused by the inflammatory disease or an element of protein losing enteropathy. Metabolic differences in fecal profiles were more marked in the CD group in comparison with the control group, indicating that the inflammation caused by CD is more extensive in comparison with UC and involves the whole intestine. Furthermore, glycerol resonances were a dominant feature of fecal spectra from patients with CD but were present in much lower intensity in the control and UC groups. This work illustrates the potential of metabonomics to generate novel noninvasive diagnostics for gastrointestinal diseases and may further our understanding of disease mechanisms.
- 137Van Immerseel, F.; Ducatelle, R.; De Vos, M.; Boon, N.; Van De Wiele, T.; Verbeke, K.; Rutgeerts, P.; Sas, B.; Louis, P.; Flint, H. J. Butyric acid-producing anaerobic bacteria as a novel probiotic treatment approach for inflammatory bowel disease. J. Med. Microbiol. 2010, 59, 141– 143, DOI: 10.1099/jmm.0.017541-0Google Scholar137https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3c%252FktVGltQ%253D%253D&md5=c0e88381cc635630a1a3d0362ffeb7c2Butyric acid-producing anaerobic bacteria as a novel probiotic treatment approach for inflammatory bowel diseaseVan Immerseel Filip; Ducatelle Richard; De Vos Martine; Boon Nico; Van De Wiele Tom; Verbeke Kristin; Rutgeerts Paul; Sas Benedikt; Louis Petra; Flint Harry JJournal of medical microbiology (2010), 59 (Pt 2), 141-143 ISSN:.There is no expanded citation for this reference.
- 138Miquel, S.; Martín, R.; Rossi, O.; Bermúdez-Humarán, L. G.; Chatel, J. M.; Sokol, H.; Thomas, M.; Wells, J. M.; Langella, P. Faecalibacterium prausnitzii and human intestinal health. Curr. Opin. Microbiol. 2013, 16, 255– 261, DOI: 10.1016/j.mib.2013.06.003Google Scholar138https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVKqsr7K&md5=9d63ee3ca46ee654075ae06da77ef52fFaecalibacterium prausnitzii and human intestinal healthMiquel, S.; Martin, R.; Rossi, O.; Bermudez-Humaran, L. G.; Chatel, J. M.; Sokol, H.; Thomas, M.; Wells, J. M.; Langella, P.Current Opinion in Microbiology (2013), 16 (3), 255-261CODEN: COMIF7; ISSN:1369-5274. (Elsevier Ltd.)A review. Faecalibacterium prausnitzii is the most abundant bacterium in the human intestinal microbiota of healthy adults, representing more than 5% of the total bacterial population. Over the past five years, an increasing no. of studies have clearly described the importance of this highly metabolically active commensal bacterium as a component of the healthy human microbiota. Changes in the abundance of F. prausnitzii have been linked to dysbiosis in several human disorders. Administration of F. prausnitzii strain A2-165 and its culture supernatant have been shown to protect against 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis in mice. Here, we discuss the role of F. prausnitzii in balancing immunity in the intestine and the mechanisms involved.
- 139Tamanai-Shacoori, Z.; Smida, I.; Bousarghin, L.; Loreal, O.; Meuric, V.; Fong, S. B.; Bonnaure-Mallet, M.; Jolivet-Gougeon, A. Roseburia spp.: a marker of health?. Future Microbiol. 2017, 12, 157– 170, DOI: 10.2217/fmb-2016-0130Google Scholar139https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslWhtbY%253D&md5=9d57c7df0d1668e2bb44e05afd2e45edRoseburia spp.: a marker of health?Tamanai-Shacoori, Zohreh; Smida, Imen; Bousarghin, Latifa; Loreal, Olivier; Meuric, Vincent; Fong, Shao Bing; Bonnaure-Mallet, Martine; Jolivet-Gougeon, AnneFuture Microbiology (2017), 12 (2), 157-170CODEN: FMUIAR; ISSN:1746-0913. (Future Medicine Ltd.)A review. The genus Roseburia consists of obligate Gram-pos. anaerobic bacteria that are slightly curved, rod-shaped and motile by means of multiple subterminal flagella. It includes five species: Roseburia intestinalis, R. hominis, R. inulinivorans, R. faecis and R. cecicola. Gut Roseburia spp. metabolize dietary components that stimulate their proliferation and metabolic activities. They are part of commensal bacteria producing short-chain fatty acids, esp. butyrate, affecting colonic motility, immunity maintenance and anti-inflammatory properties. Modification in Roseburia spp. representation may affect various metabolic pathways and is assocd. with several diseases (including irritable bowel syndrome, obesity, Type 2 diabetes, nervous system conditions and allergies). Roseburia spp. could also serve as biomarkers for symptomatic pathologies (e.g., gallstone formation) or as probiotics for restoration of beneficial flora.
- 140Li, Y.; Xia, S.; Jiang, X.; Feng, C.; Gong, S.; Ma, J.; Fang, Z.; Yin, J.; Yin, Y. Gut Microbiota and Diarrhea: An Updated Review. Microbiology 2021, 11, 625210, DOI: 10.3389/fcimb.2021.625210Google Scholar140https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3sbpt1aluw%253D%253D&md5=efb07829392f2514713f0ede26ef97f8Gut Microbiota and Diarrhea: An Updated ReviewLi Yunxia; Xia Siting; Jiang Xiaohan; Feng Can; Gong Saiming; Ma Jie; Yin Jie; Yin Yulong; Fang Zhengfeng; Yin YulongFrontiers in cellular and infection microbiology (2021), 11 (), 625210 ISSN:.Diarrhea is a common problem to the whole world and the occurrence of diarrhea is highly associated with gut microbiota, such as bacteria, fungi, and viruses. Generally, diarrheal patients or animals are characterized by gut microbiota dysbiosis and pathogen infections may lead to diarrheal phenotypes. Of relevance, reprograming gut microbiota communities by dietary probiotics or fecal bacteria transplantation are widely introduced to treat or prevent diarrhea. In this review, we discussed the influence of the gut microbiota in the infection of diarrhea pathogens, and updated the research of reshaping the gut microbiota to prevent or treat diarrhea for the past few years. Together, gut microbiota manipulation is of great significance to the prevention and treatment of diarrhea, and further insight into the function of the gut microbiota will help to discover more anti-diarrhea probiotics.
- 141Hodges, K.; Gill, R. Infectious diarrhea: Cellular and molecular mechanisms. Gut Microbes 2010, 1, 4– 21, DOI: 10.4161/gmic.1.1.11036Google Scholar141https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2sboslOhtA%253D%253D&md5=b0992f6a97ff50dff635deea9065d231Infectious diarrhea: Cellular and molecular mechanismsHodges Kim; Gill RavinderGut microbes (2010), 1 (1), 4-21 ISSN:.Diarrhea caused by enteric infections is a major factor in morbidity and mortality worldwide. An estimated 2-4 billion episodes of infectious diarrhea occur each year and are especially prevalent in infants. This review highlights the cellular and molecular mechanisms underlying diarrhea associated with the three classes of infectious agents, i.e., bacteria, viruses and parasites. Several bacterial pathogens have been chosen as model organisms, including Vibrio cholerae as a classical example of secretory diarrhea, Clostridium difficile and Shigella species as agents of inflammatory diarrhea and selected strains of pathogenic Escherichia coli (E. coli) to discuss the recent advances in alteration of epithelial ion absorption. Many of the recent studies addressing epithelial ion transport and barrier function have been carried out using viruses and parasites. Here, we focus on the rapidly developing field of viral diarrhea including rotavirus, norovirus and astrovirus infections. Finally we discuss Giardia lamblia and Entamoeba histolytica as examples of parasitic diarrhea. Parasites have a greater complexity than the other pathogens and are capable of creating molecules similar to those produced by the host, such as serotonin and PGE(2). The underlying mechanisms of infectious diarrhea discussed include alterations in ion transport and tight junctions as well as the virulence factors, which alter these processes either through direct effects or indirectly through inflammation and neurotransmitters.
- 142Levine, M. M. Escherichia coli that cause diarrhea: enterotoxigenic, enteropathogenic, enteroinvasive, enterohemorrhagic, and enteroadherent. J. Infect. Dis. 1987, 155, 377– 389, DOI: 10.1093/infdis/155.3.377Google Scholar142https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL2s7htVylsw%253D%253D&md5=28f3bb6db9138f13a131fadf174fe710Escherichia coli that cause diarrhea: enterotoxigenic, enteropathogenic, enteroinvasive, enterohemorrhagic, and enteroadherentLevine M MThe Journal of infectious diseases (1987), 155 (3), 377-89 ISSN:0022-1899.There are four major categories of diarrheagenic Escherichia coli: enterotoxigenic (a major cause of travelers' diarrhea and infant diarrhea in less-developed countries), enteroinvasive (a cause of dysentery), enteropathogenic (an important cause of infant diarrhea), and enterohemorrhagic (a cause of hemorrhagic colitis and hemolytic uremic syndrome). Besides manifesting distinct clinical patterns, these categories of E. coli differ in their epidemiology and pathogenesis and in their O:H serotypes. Common features (albeit distinct for each category) include plasmid-encoded virulence properties, characteristic interactions with intestinal mucosa, and elaboration of various types of enterotoxins or cytotoxins. A less-well-defined fifth category of diarrheagenic E. coli is that of enteroadherent E. coli, so far identifiable only by their pattern of adherence to Hep-2 cells in tissue culture.
- 143Gallo, A.; Passaro, G.; Gasbarrini, A.; Landolfi, R.; Montalto, M. Modulation of microbiota as treatment for intestinal inflammatory disorders: An uptodate. World J. Gastroenterol. 2016, 22, 7186– 7202, DOI: 10.3748/wjg.v22.i32.7186Google Scholar143https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2svgtVCqtw%253D%253D&md5=976dc72a198f8a598627eb8a347d3c7cModulation of microbiota as treatment for intestinal inflammatory disorders: An uptodateGallo Antonella; Passaro Giovanna; Gasbarrini Antonio; Landolfi Raffaele; Montalto MassimoWorld journal of gastroenterology (2016), 22 (32), 7186-202 ISSN:.Alterations of intestinal microflora may significantly contribute to the pathogenesis of different inflammatory and autoimmune disorders. There is emerging interest on the role of selective modulation of microflora in inducing benefits in inflammatory intestinal disorders, by as probiotics, prebiotics, synbiotics, antibiotics, and fecal microbiota transplantation (FMT). To summarize recent evidences on microflora modulation in main intestinal inflammatory disorders, PubMed was searched using terms microbiota, intestinal flora, probiotics, prebiotics, fecal transplantation. More than three hundred articles published up to 2015 were selected and reviewed. Randomized placebo-controlled trials and meta-analysis were firstly included, mainly for probiotics. A meta-analysis was not performed because of the heterogeneity of these studies. Most of relevant data derived from studies on probiotics, reporting some efficacy in ulcerative colitis and in pouchitis, while disappointing results are available for Crohn's disease. Probiotic supplementation may significantly reduce rates of rotavirus diarrhea. Efficacy of probiotics in NSAID enteropathy and irritable bowel syndrome is still controversial. Finally, FMT has been recently recognized as an efficacious treatment for recurrent Clostridium difficile infection. Modulation of intestinal flora represents a very interesting therapeutic target, although it still deserves some doubts and limitations. Future studies should be encouraged to provide new understanding about its therapeutical role.
- 144Bron, P. A.; Kleerebezem, M.; Brummer, R. J.; Cani, P. D.; Mercenier, A.; MacDonald, T. T.; Garcia-Ródenas, C. L.; Wells, J. M. Can probiotics modulate human disease by impacting intestinal barrier function?. Br. J. Nutr. 2017, 117, 93– 107, DOI: 10.1017/S0007114516004037Google Scholar144https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvVyks74%253D&md5=73dd9acb7e2cf433fbcfb1a3d706d8e5Can probiotics modulate human disease by impacting intestinal barrier function?Bron, Peter A.; Kleerebezem, Michiel; Brummer, Robert-Jan; Cani, Patrice D.; Mercenier, Annick; MacDonald, Thomas T.; Garcia-Rodenas, Clara L.; Wells, Jerry M.British Journal of Nutrition (2017), 117 (1), 93-107CODEN: BJNUAV; ISSN:0007-1145. (Cambridge University Press)Intestinal barrier integrity is a prerequisite for homeostasis of mucosal function, which is balanced to maximise absorptive capacity, while maintaining efficient defensive reactions against chem. and microbial challenges. Evidence is mounting that disruption of epithelial barrier integrity is one of the major etiol. factors assocd. with several gastrointestinal diseases, including infection by pathogens, obesity and diabetes, necrotising enterocolitis, irritable bowel syndrome and inflammatory bowel disease. The notion that specific probiotic bacterial strains can affect barrier integrity fuelled research in which in vitro cell lines, animal models and clin. trials are used to assess whether probiotics can revert the diseased state back to homeostasis and health. This review catalogues and categorises the lines of evidence available in literature for the role of probiotics in epithelial integrity and, consequently, their beneficial effect for the redn. of gastrointestinal disease symptoms.
- 145Zhang, S.; Wang, R.; Li, D.; Zhao, L.; Zhu, L. Role of gut microbiota in functional constipation. Gastroenterol. Rep. 2021, 9, 392– 401, DOI: 10.1093/gastro/goab035Google Scholar145https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2cjos1eitw%253D%253D&md5=d9df2a9e9bd37ff2920138061a0d473eRole of gut microbiota in functional constipationZhang Shengsheng; Wang Ruixin; Li Danyan; Zhao Luqing; Zhu LixinGastroenterology report (2021), 9 (5), 392-401 ISSN:2052-0034.Functional constipation (FC) is common, yet the etiology is not clear. Accumulating evidence suggests an association between FC and abnormal gut microbiota. The relationship between the gut microbiota and the gut transit is likely bidirectional. This review summarizes the current evidence regarding the impact of gut microbiota on the pathogenesis of FC. By modulating the colonic motility, secretion, and absorption, gut microbiota may contribute to the development of FC through microbial metabolic activities involving bile acids, short-chain fatty acids, 5-hydroxytryptamine, and methane. In support of the key roles of the gut microbiota in FC, treatment with probiotics, prebiotics, synbiotics, and traditional Chinese medicine often result in compositional and functional changes in the gut microbiota. Further studies on the pathogenesis of FC and the therapeutic mechanism of microecological agents will provide a knowledge base for better management of FC.
- 146Zoppi, G.; Cinquetti, M.; Luciano, A.; Benini, A.; Muner, A.; Bertazzoni Minelli, E. The intestinal ecosystem in chronic functional constipation. Acta Paediatr. 1998, 87, 836– 841, DOI: 10.1111/j.1651-2227.1998.tb01547.xGoogle Scholar146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK1cvgsFKgtQ%253D%253D&md5=bb92c7610207657d2e9c5dad125d53c2The intestinal ecosystem in chronic functional constipationZoppi G; Cinquetti M; Luciano A; Benini A; Muner A; Bertazzoni Minelli EActa paediatrica (Oslo, Norway : 1992) (1998), 87 (8), 836-41 ISSN:0803-5253.Chronic functional constipation is common in infants, and the bacterial composition of stools in this condition is not known. The study aims were to: (i) investigate the composition of the intestinal ecosystem in chronic functional constipation; (ii) establish whether the addition of the water-holding agent calcium polycarbophil to the diet induces an improvement in constipation; and (iii) determine the composition of the intestinal ecosystem after the use of this agent. In total, 42 children (20F, 22M; mean age: 8.6 +/- 2.9 y) were studied. Twenty-eight children with functional chronic constipation without anatomical disorders were treated double-blind in random sequence for 1 month with an oral preparation of calcium polycarbophil (0.62 g/twice daily) or placebo. Intestinal flora composition was evaluated by standard microbiological methods and biochemical assays on faecal samples collected before and after treatment. Fourteen healthy children were studied as controls. The results show that (i) the constipated children presented a significant increase in clostridia and bifidobacteria in faeces compared to healthy subjects--different species of clostridia and enterobacteriaceae were frequently isolated; no generalized overgrowth was observed; Clostridia outnumbered bacteroides and E. coli mean counts by 2-3log, while bacteroides and E. coli counts were similar (5-6 log10/g fresh faeces); these intestinal disturbances could be defined as a dysbiosis, i.e. a quantitative alteration in the relative proportions of certain intestinal bacterial species. (ii) Clinical resolution of constipation was achieved only in 43% of treated children and an improvement in 21% (one bowel movement every 2 d). (iii) Calcium polycarbophil treatment induced no significant changes in the composition of the intestinal ecosystem, nor in blood chemistry parameters.
- 147Khalif, I. L.; Quigley, E. M.; Konovitch, E. A.; Maximova, I. D. Alterations in the colonic flora and intestinal permeability and evidence of immune activation in chronic constipation. Dig. Liver Dis. 2005, 37, 838– 849, DOI: 10.1016/j.dld.2005.06.008Google Scholar147https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2MrmsFCntQ%253D%253D&md5=7b2a49da0e98a1645fa10ad657392e4cAlterations in the colonic flora and intestinal permeability and evidence of immune activation in chronic constipationKhalif I L; Quigley E M M; Konovitch E A; Maximova I DDigestive and liver disease : official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver (2005), 37 (11), 838-49 ISSN:1590-8658.BACKGROUND: Disturbances in bowel function in chronic constipation could result in changes in the colonic flora and lead to disordered immunity and to decreased resistance to pathogenic flora. AIM: To investigate systemic immunity, the faecal flora and intestinal permeability in patients with chronic constipation, under basal conditions and following therapy with the laxative Bisacodyl. METHODS: Intestinal permeability, faecal flora analysis, T- and B-lymphocyte numbers, T-cell subpopulations, lymphocyte proliferation, phagocytosis, intracellular killing of Staphylococcus aureus by neutrophils, as well as circulating levels of immunoglobulins, immune complexes and antibacterial antibodies were assessed in 57 patients with functional constipation. In 12 patients with severely delayed transit, investigations were repeated following therapy with Bisacodyl. RESULTS: Ovalbumin concentrations, in serum, were higher in constipated patients (28.2+/-4.1 ng/ml versus 1.0+/-0.4 ng/ml, p < 0.05). Elevated counts of CD3+, CD4+, CD25+ cells, increased spontaneous proliferation of lymphocytes, elevated titres of antibodies to Escherichia coli and S. aureus, diminished counts of CD72+ B cells, diminished lymphocyte proliferation under phytohemagglutinin (PHA) stimulation and a diminished phagocytic index for both neutrophils and monocytes were found in the constipated patients. Concentrations of Bifidobacterium and Lactobacillus were significantly lower in constipated patients; potentially pathogenic bacteria and/or fungi were increased. Therapy with Bisacodyl resulted in normalisation of the faecal flora, a reduction in ovalbumin concentration and return towards normal for certain immunologic parameters. CONCLUSION: Constipation is associated with striking changes in the faecal flora, intestinal permeability and the systemic immune response. Relief of constipation tends to normalise these findings suggesting that these changes are secondary to, rather than a cause of, constipation.
- 148Kim, S. E.; Choi, S. C.; Park, K. S.; Park, M. I.; Shin, J. E.; Lee, T. H.; Jung, K. W.; Koo, H. S.; Myung, S. J. Change of Fecal Flora and Effectiveness of the Short-term VSL#3 Probiotic Treatment in Patients With Functional Constipation. J. Neurogastroenterol. Motil. 2015, 21, 111– 120, DOI: 10.5056/jnm14048Google Scholar148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MvhslOltA%253D%253D&md5=bfd9824ac1d168c6ba1dbc0aa8172459Change of Fecal Flora and Effectiveness of the Short-term VSL#3 Probiotic Treatment in Patients With Functional ConstipationKim Seong-Eun; Choi Suck Chei; Park Kyung Sik; Park Moo In; Shin Jeong Eun; Lee Tae Hee; Jung Kee Wook; Myung Seung-Jae; Koo Hoon SupJournal of neurogastroenterology and motility (2015), 21 (1), 111-20 ISSN:2093-0879.BACKGROUND/AIMS: We investigated gut flora characteristics in patients with functional constipation (FC) and influences of short-term treatment with VSL#3 probiotic on flora and symptom improvement. METHODS: Thirty patients fulfilling Rome III criteria for FC and 30 controls were enrolled. Fecal samples were obtained before and after VSL#3 intake (one sachet twice daily for 2 weeks) and flora were examined by quantitative real-time polymerase chain reaction (qRT-PCR). Symptom changes were also investigated. RESULTS: The fold differences in Bifidobacterium and Bacteroides species were significantly lower in feces from FC, compared to in controls (P = 0.030 and P = 0.021). After taking VSL#3, the fold differences in Lactobacillus, Bifidobacterium and Bacteroides species increased in controls (P = 0.022, P = 0.018, and P = 0.076), but not in FC. Mean Bristol scores and complete spontaneous bowel movements (CSBMs)/week increased significantly in FC after ingesting VSL#3 (both P < 0.001). Relief of subjective CSBM frequency, stool consistency and abdominal bloating were reported in 70%, 60%, and 47% of patients. After VSL#3 cessation, 44.4% of patients with symptom improvement experienced constipation recurrence mostly within one month. CONCLUSIONS: Bifidobacterium and Bacteroides species might be quantitatively altered in FC. A short-term VSL#3 treatment can improve clinical symptoms of FC. Further studies are needed to investigate VSL#3's additional effects beyond altering gut flora to allevate constipation.
- 149Zhu, L.; Liu, W.; Alkhouri, R.; Baker, R. D.; Bard, J. E.; Quigley, E. M.; Baker, S. S. Structural changes in the gut microbiome of constipated patients. Physiol. Genomics 2014, 46, 679– 686, DOI: 10.1152/physiolgenomics.00082.2014Google Scholar149Structural changes in the gut microbiome of constipated patientsZhu, Lixin; Liu, Wensheng; Alkhouri, Razan; Baker, Robert D.; Bard, Jonathan E.; Quigley, Eamonn M.; Baker, Susan S.Physiological Genomics (2014), 46 (18), 679-686CODEN: PHGEFP; ISSN:1094-8341. (American Physiological Society)Previous studies using culture-based methods suggested an assocn. between constipation and altered abundance of certain taxa of the colonic microbiome. We aim to examine the global changes in gut microbial compn. of constipated patients. A cross-sectional pilot study using 16S rRNA gene pyrosequencing was performed to compare stool microbial compn. of eight constipated patients and 14 nonconstipated controls. Only obes