Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 299
Filter
Add more filters

Publication year range
1.
J Periodontal Res ; 59(4): 758-770, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38699835

ABSTRACT

BACKGROUND AND OBJECTIVE: Osteoporosis is associated with bone microarchitecture alterations, and the depletion of estrogen during menopause is a major contributing factor to its development. The literature highlights the noteworthy role of gut microbiota in bone metabolism, particularly in the progression of osteoporosis. Periodontal disease leads to alveolar bone loss, which may be influenced by estrogen deficiency, and this mechanism is intricately associated with an imbalance in systemic microbiota. The aim of this study was to evaluate the effects of Bifidobacterium animalis subsp. lactis HN019 (B. lactis HN019) and Lacticaseibacillus casei 01 (L. casei 01) administrations on an osteoporosis animal model. MATERIALS AND METHODS: Thirty-three female rats were randomly divided into three groups: control (C-OVX), C-OVX-HN019 and C-OVX-LC01. All animals were ovariectomized. In groups C-OVX-HN019 and C-OVX-LC01, the probiotics were administered for 4 months. All animals were euthanized after 16 weeks from ovariectomy. Microtomographic, histopathological and immunohistochemical examinations were conducted on periodontal tissues, whereas histomorphometry, histopathological and immunohistochemical analyses were carried out on the intestine. The levels of estradiol were assessed in blood using an immunoenzymatic assay. The data were subjected to statistical analyses (p < .05). RESULTS: The C-OVX-LC01 group exhibited a significant reduction in alveolar bone porosity and an increase in connective tissue density compared to C-OVX (p < .05). The C-OVX-HN019 and C-OVX-LC01 groups presented reduced expression of TRAP and RANKL compared to the C-OVX (p < .05). The C-OVX group presented villi defects, mild neutrophil infiltration, decrease in both villous height and intestinal crypts and reduced expression of intestinal junctional epithelium markers e-cadherin and claudin 01 compared to C-OVX-HN019 and C-OVX-LC01 (p < .05). The C-OVX group had lower estradiol levels than C-OVX-HN019 and C-OVX-LC01 (p < .05). CONCLUSION: The probiotic therapy promoted a reduction in alveolar bone destruction and intestinal permeability as well as an increase in estradiol levels in ovariectomized rats. Specifically, the probiotic strain Lacticaseibacillus casei 01 exhibited greater effectiveness compared to Bifidobacterium animalis subsp. lactis HN019, indicating strain-dependent outcomes.


Subject(s)
Estradiol , Osteoporosis , Ovariectomy , Probiotics , Animals , Estradiol/blood , Probiotics/therapeutic use , Probiotics/pharmacology , Female , Rats , Osteoporosis/pathology , Alveolar Bone Loss/pathology , Alveolar Bone Loss/prevention & control , Disease Models, Animal , Lacticaseibacillus casei , Bifidobacterium animalis , X-Ray Microtomography , Alveolar Process/pathology , Intestines/pathology , Intestines/microbiology , Gastrointestinal Microbiome , Rats, Wistar
2.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Article in English | MEDLINE | ID: mdl-34610963

ABSTRACT

The microbial communities in the mouth and colon are anatomically connected via the saliva. However, the extent to which oral microbes reach and successfully colonize the distal gut has been debated. To resolve this long-standing controversy, we used exact amplicon sequence variants generated from concurrently collected saliva/stool microbiota in 66 healthy adults from two countries to show that, with one exception (Dialister invisus), the two niches are completely distinct. Thus, there is no evidence for colonization of oral bacteria in the distal gut. This defines the healthy state to which pathological states could be compared. Finding the same bacteria in the mouth and stool may warrant clinical investigation for an underlying pathology.


Subject(s)
Bacteria/growth & development , Intestines/microbiology , Mouth/microbiology , Adult , Bacteria/classification , Feces/microbiology , Gastrointestinal Microbiome , Humans , Phylogeny
3.
J Fish Biol ; 104(6): 2068-2080, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38596840

ABSTRACT

The pervasive utilization of plastics and their integration into ecosystems has resulted in significant environmental issues, particularly the pollution of microplastics (MPs). In aquaculture, high-fat feed (HFD) is frequently employed to enhance the energy intake and economic fish production. This study utilized zebrafish as a model organism to investigate the impact of concurrent exposure to HFD and MPs on fish intestinal pathology damage and intestinal microbiome. The experimental design involved the division of zebrafish into two groups: one receiving a normal diet (ND) and the other receiving HFD. The zebrafish were exposed to a control group, as well as polystyrene (PS) MPs of varying sizes (5 and 50 µm). Histopathological examination revealed that the combination of 5 µm MPs and HFD resulted in the most significant damage to the zebrafish intestinal tract. Furthermore, gut microbiome assays indicated that exposure to MPs and HFD altered the composition of the gut microbiome. This study demonstrates that in aquaculture, the issue of HFD must be considered alongside concerns about MPs contamination, as both factors appear to have a combined effect on the intestinal pathology damage and intestinal microbiome. The findings of this research offer valuable insights for the improvement of fish farming practices.


Subject(s)
Gastrointestinal Microbiome , Intestines , Microplastics , Polystyrenes , Water Pollutants, Chemical , Zebrafish , Animals , Zebrafish/microbiology , Microplastics/toxicity , Polystyrenes/toxicity , Polystyrenes/adverse effects , Gastrointestinal Microbiome/drug effects , Intestines/pathology , Intestines/microbiology , Intestines/drug effects , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/adverse effects , Aquaculture , Diet, High-Fat/adverse effects , Animal Feed/analysis
4.
Nature ; 544(7650): 357-361, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28273061

ABSTRACT

Recent genomic data have revealed multiple interactions between Neanderthals and modern humans, but there is currently little genetic evidence regarding Neanderthal behaviour, diet, or disease. Here we describe the shotgun-sequencing of ancient DNA from five specimens of Neanderthal calcified dental plaque (calculus) and the characterization of regional differences in Neanderthal ecology. At Spy cave, Belgium, Neanderthal diet was heavily meat based and included woolly rhinoceros and wild sheep (mouflon), characteristic of a steppe environment. In contrast, no meat was detected in the diet of Neanderthals from El Sidrón cave, Spain, and dietary components of mushrooms, pine nuts, and moss reflected forest gathering. Differences in diet were also linked to an overall shift in the oral bacterial community (microbiota) and suggested that meat consumption contributed to substantial variation within Neanderthal microbiota. Evidence for self-medication was detected in an El Sidrón Neanderthal with a dental abscess and a chronic gastrointestinal pathogen (Enterocytozoon bieneusi). Metagenomic data from this individual also contained a nearly complete genome of the archaeal commensal Methanobrevibacter oralis (10.2× depth of coverage)-the oldest draft microbial genome generated to date, at around 48,000 years old. DNA preserved within dental calculus represents a notable source of information about the behaviour and health of ancient hominin specimens, as well as a unique system that is useful for the study of long-term microbial evolution.


Subject(s)
DNA, Ancient/analysis , Dental Calculus/chemistry , Diet/history , Food Preferences , Health/history , Neanderthals/microbiology , Neanderthals/psychology , Animals , Belgium , Carnivory , Caves , Enterocytozoon/genetics , Enterocytozoon/isolation & purification , Genome, Bacterial/genetics , History, Ancient , Humans , Intestines/microbiology , Meat/history , Methanobrevibacter/genetics , Methanobrevibacter/isolation & purification , Mouth/microbiology , Pan troglodytes/microbiology , Penicillium/chemistry , Perissodactyla , Sheep , Spain , Stomach/microbiology , Symbiosis , Time Factors , Vegetarians/history
5.
Int J Mol Sci ; 24(3)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36768285

ABSTRACT

Intestinal dysbiosis is related to the physiopathology and clinical manifestation of rheumatoid arthritis (RA) and the response to pharmacologic treatment. The objectives of this study were (1) to analyze the effect of conventional synthetic disease modifying anti-rheumatic drugs (csDMARDs) on the abundance of gut microbiota's bacteria; (2) to evaluate the relationship between the differences in microbial abundance with the serum levels of intestinal fatty-acid binding protein 2 (IFABP2), cytokines, and the response phenotype to csDMARDs therapy in RA. A cross-sectional study was conducted on 23 women diagnosed with RA. The abundance of bacteria in gut microbiota was determined with qPCR. The ELISA technique determined serum levels of IFABP2, TNF-α, IL-10, and IL-17A. We found that the accumulated dose of methotrexate or prednisone is negatively associated with the abundance of Lactobacillus but positively associated with the abundance of Bacteroides fragilis. The Lactobacillus/Porphyromonas gingivalis ratio was associated with the Disease Activity Score-28 for RA with Erythrocyte Sedimentation Rate (DAS28-ESR) (r = 0.778, p = 0.030) and with the levels of IL-17A (r = 0.785, p = 0.027) in the group treated with csDMARD. Moreover, a relation between the serum levels of IFABP2 and TNF-α (r = 0.593, p = 0.035) was observed in the group treated with csDMARD. The serum levels of IFABP2 were higher in patients with secondary non-response to csDMARDs therapy. In conclusion, our results suggest that the ratios of gut microbiota's bacteria and intestinal permeability seems to establish the preamble for therapeutic secondary non-response in RA.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Gastrointestinal Microbiome , Lactobacillus , Female , Humans , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Cross-Sectional Studies , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Interleukin-17 , Pilot Projects , Porphyromonas gingivalis , Tumor Necrosis Factor-alpha/therapeutic use , Intestines/microbiology , Intestines/physiopathology , Cell Membrane Permeability
6.
Nature ; 534(7606): 259-62, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27279224

ABSTRACT

Human-associated microbial communities have a crucial role in determining our health and well-being, and this has led to the continuing development of microbiome-based therapies such as faecal microbiota transplantation. These microbial communities are very complex, dynamic and highly personalized ecosystems, exhibiting a high degree of inter-individual variability in both species assemblages and abundance profiles. It is not known whether the underlying ecological dynamics of these communities, which can be parameterized by growth rates, and intra- and inter-species interactions in population dynamics models, are largely host-independent (that is, universal) or host-specific. If the inter-individual variability reflects host-specific dynamics due to differences in host lifestyle, physiology or genetics, then generic microbiome manipulations may have unintended consequences, rendering them ineffective or even detrimental. Alternatively, microbial ecosystems of different subjects may exhibit universal dynamics, with the inter-individual variability mainly originating from differences in the sets of colonizing species. Here we develop a new computational method to characterize human microbial dynamics. By applying this method to cross-sectional data from two large-scale metagenomic studies--the Human Microbiome Project and the Student Microbiome Project--we show that gut and mouth microbiomes display pronounced universal dynamics, whereas communities associated with certain skin sites are probably shaped by differences in the host environment. Notably, the universality of gut microbial dynamics is not observed in subjects with recurrent Clostridium difficile infection but is observed in the same set of subjects after faecal microbiota transplantation. These results fundamentally improve our understanding of the processes that shape human microbial ecosystems, and pave the way to designing general microbiome-based therapies.


Subject(s)
Ecosystem , Microbiota/physiology , Clostridioides difficile/physiology , Clostridium Infections/microbiology , Computer Simulation , Cross-Sectional Studies , Datasets as Topic , Environment , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , Healthy Volunteers , Humans , Intestines/microbiology , Metagenomics , Mouth/microbiology , Organ Specificity , Skin/microbiology , Species Specificity
7.
J Pediatr ; 228: 110-116.e1, 2021 01.
Article in English | MEDLINE | ID: mdl-32918918

ABSTRACT

OBJECTIVE: To identify exposure pathways to fecal pathogens that are significant contributors to diarrheal diseases and impaired growth in young children, and to evaluate scalable interventions to reduce fecal contamination from these pathways. STUDY DESIGN: Reducing Enteropathy, Undernutrition, and Contamination in the Environment (REDUCE) was a prospective cohort study of 370 children <5 years of age was conducted in Walungu Territory, South Kivu, Democratic Republic of the Congo. Child mouthing behaviors were assessed through caregiver reports and 5-hour structured observations. Caregiver reports of child contact with animals and child diarrhea were also obtained. Anthropometric measurements were collected at baseline and at a 6-month follow-up. RESULTS: Children observed putting soil in their mouth during structured observation at baseline had a significantly higher odds of diarrhea at the 6-month follow-up (OR, 1.79; 95% CI, 1.04 to 3.07). Children observed mouthing feces during structured observation had a significant reduction in height-for-age z-score (HAZ) from baseline to the 6-month follow-up (ΔHAZ, -0.69; 95% CI, -1.34 to -0.04). A significant reduction in HAZ was also observed for children with caregiver reports of touching guinea pigs (-0.33; 95% CI, -0.58 to -0.08) and rabbits (-0.34; 95% CI, -0.64 to -0.04) and children with feces in their sleeping space during unannounced spot checks (-0.41; 95% CI, -0.74 to -0.09). CONCLUSIONS: These findings emphasize the urgent need for infant water, sanitation, and hygiene interventions targeting child mouthing behaviors, fecal contamination in child living spaces, and child contact with domestic animals to reduce exposure to fecal pathogens among susceptible populations.


Subject(s)
Child Behavior , Diarrhea/epidemiology , Fomites/microbiology , Hygiene , Malnutrition/epidemiology , Animals , Child, Preschool , Congo/epidemiology , Diarrhea/etiology , Diarrhea/prevention & control , Environmental Exposure/adverse effects , Feces , Female , Follow-Up Studies , Guinea Pigs , Humans , Infant , Infant, Newborn , Intestines/microbiology , Male , Mouth , Prospective Studies , Rabbits
8.
Gastroenterology ; 156(4): 1041-1051.e4, 2019 03.
Article in English | MEDLINE | ID: mdl-30445014

ABSTRACT

BACKGROUND & AIMS: Bile diversion to the ileum (GB-IL) has strikingly similar metabolic and satiating effects to Roux-en-Y gastric bypass (RYGB) in rodent obesity models. The metabolic benefits of these procedures are thought to be mediated by increased bile acids, although parallel changes in body weight and other confounding variables limit this interpretation. METHODS: Global G protein-coupled bile acid receptor-1 null (Tgr5-/-) and intestinal-specific farnesoid X receptor null (FxrΔ/E) mice on high-fat diet as well as wild-type C57BL/6 and glucagon-like polypeptide 1 receptor deficient (Glp-1r-/-) mice on chow diet were characterized following GB-IL. RESULTS: GB-IL induced weight loss and improved oral glucose tolerance in Tgr5-/-, but not FxrΔ/E mice fed a high-fat diet, suggesting a role for intestinal Fxr. GB-IL in wild-type, chow-fed mice prompted weight-independent improvements in glycemia and glucose tolerance secondary to augmented insulin responsiveness. Improvements were concomitant with increased levels of lymphatic GLP-1 in the fasted state and increased levels of intestinal Akkermansia muciniphila. Improvements in fasting glycemia after GB-IL were mitigated with exendin-9, a GLP-1 receptor antagonist, or cholestyramine, a bile acid sequestrant. The glucoregulatory effects of GB-IL were lost in whole-body Glp-1r-/- mice. CONCLUSIONS: Bile diversion to the ileum improves glucose homeostasis via an intestinal Fxr-Glp-1 axis. Altered intestinal bile acid availability, independent of weight loss, and intestinal Akkermansia muciniphila appear to mediate the metabolic changes observed after bariatric surgery and might be manipulated for treatment of obesity and diabetes.


Subject(s)
Bile Acids and Salts/metabolism , Blood Glucose/metabolism , Gallbladder/surgery , Glucagon-Like Peptide 1/metabolism , Ileum/surgery , Receptors, Cytoplasmic and Nuclear/metabolism , Anastomosis, Surgical , Animals , Anticholesteremic Agents/pharmacology , Bariatric Surgery , Cholestyramine Resin/pharmacology , Diet, High-Fat , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/genetics , Glucose Tolerance Test , Insulin Resistance , Intestines/microbiology , Lymph/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Verrucomicrobia , Weight Loss
9.
Microb Cell Fact ; 19(1): 50, 2020 Feb 27.
Article in English | MEDLINE | ID: mdl-32103766

ABSTRACT

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, and in an effort to identify novel therapeutic target for this disease in recent years, human microbiota has attracted much interest. This paper briefly summarizes the main findings concerning the differences of human microbiome across several important mucosal interfaces, including nose, mouth, and gut between PD patients and controls as obtained from a total of 13 studies published since 2015, which covered a total of 943 PD patients and 831 matched controls from 6 countries. Overall, these studies supported the differences of gut microbiota between PD patients and matched controls, while significantly altered bacterial taxa among studies were not identical. Due to relatively limited number of available studies and covered patients, the associations between oral and nasal microbiota and PD remain inconclusive. The therapeutic and diagnostic potentials of gut microbiota for PD are discussed. More well-designed clinical studies recruiting large-scale PD patients are encouraged in future.


Subject(s)
Intestines/microbiology , Microbiota , Mouth/microbiology , Nose/microbiology , Parkinson Disease/microbiology , Humans
10.
Microb Cell Fact ; 19(1): 216, 2020 Nov 26.
Article in English | MEDLINE | ID: mdl-33243224

ABSTRACT

BACKGROUND: Gut microbiota in humans and animals play an important role in health, aiding in digestion, regulation of the immune system and protection against pathogens. Changes or imbalances in the gut microbiota (dysbiosis) have been linked to a variety of local and systemic diseases, and there is growing evidence that restoring the balance of the microbiota by delivery of probiotic microorganisms can improve health. However, orally delivered probiotic microorganisms must survive transit through lethal highly acid conditions of the stomach and bile salts in the small intestine. Current methods to protect probiotic microorganisms are still not effective enough. RESULTS: We have developed a cell encapsulation technology based on the natural polymer, cellulose sulphate (CS), that protects members of the microbiota from stomach acid and bile. Here we show that six commonly used probiotic strains (5 bacteria and 1 yeast) can be encapsulated within CS microspheres. These encapsulated strains survive low pH in vitro for at least 4 h without appreciable loss in viability as compared to their respective non-encapsulated counterparts. They also survive subsequent exposure to bile. The CS microspheres can be digested by cellulase at concentrations found in the human intestine, indicating one mechanism of release. Studies in mice that were fed CS encapsulated autofluorescing, commensal E. coli demonstrated release and colonization of the intestinal tract. CONCLUSION: Taken together, the data suggests that CS microencapsulation can protect bacteria and yeasts from viability losses due to stomach acid, allowing the use of lower oral doses of probiotics and microbiota, whilst ensuring good intestinal delivery and release.


Subject(s)
Cell Encapsulation/methods , Cellulose/analogs & derivatives , Drug Compounding/methods , Drug Delivery Systems/methods , Escherichia coli/growth & development , Probiotics/administration & dosage , Animals , Cellulase/chemistry , Cellulose/chemistry , Gastric Juice , Gastrointestinal Microbiome , Humans , Hydrogen-Ion Concentration , Intestines/microbiology , Male , Mice , Mice, Nude , Microbial Viability , Microspheres
11.
Neuropathol Appl Neurobiol ; 45(3): 195-215, 2019 04.
Article in English | MEDLINE | ID: mdl-30298527

ABSTRACT

Inflammation and neurodegeneration are key features of many chronic neurological diseases, yet the causative mechanisms underlying these processes are poorly understood. There has been mounting interest in the role of the human microbiome in modulating the inflammatory milieu of the central nervous system (CNS) in health and disease. To date, most research has focussed on a gut-brain axis, with other mucosal surfaces being relatively neglected. We herein take the novel approach of comprehensively reviewing the roles of the microbiome across several key mucosal interfaces - the nose, mouth, lung and gut - in health and in Parkinson's disease (PD), Alzheimer's disease (AD) and multiple sclerosis (MS). This review systematically appraises the anatomical and microbiological landscape of each mucosal surface in health and disease before considering relevant mechanisms that may influence the initiation and progression of PD, AD and MS. The cumulative effects of dysbiosis from the nose to the gut may contribute significantly to neurological disease through a wide variety of mechanisms, including direct translocation of bacteria and their products, and modulation of systemic or CNS-specific immunity. This remains an understudied and exciting area for future research and may lead to the development of therapeutic targets for chronic neurological disease.


Subject(s)
Alzheimer Disease/microbiology , Dysbiosis/microbiology , Inflammation/microbiology , Intestines/microbiology , Lung/microbiology , Microbiota , Mouth/microbiology , Multiple Sclerosis/microbiology , Nasal Cavity/microbiology , Olfaction Disorders/microbiology , Parkinson Disease/microbiology , Alzheimer Disease/complications , Humans , Multiple Sclerosis/complications , Olfaction Disorders/etiology , Parkinson Disease/complications
12.
Fish Shellfish Immunol ; 91: 251-263, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31121290

ABSTRACT

Soiny mullet (Liza haematocheila) is an important economic fish species in China, but stress and diseases have seriously restricted its culture. There are no effective methods including vaccines to prevent or control these diseases. Alternative methods should be employed, such as using novel immunostimulant poly-ß-hydroxybutyrate (PHB). The present study aimed to evaluate effects of dietary PHB supplementation on the growth, antioxidant enzymes activity, immune-related genes expression and intestinal microbiota in soiny mullet. The fish was fed for 30 or 60 days with six diets at different PHB supplementation of 0, 0.5, 1, 2, 4 and 8%, named as groups P0, P0.5, P1, P2, P4 and P8. The results showed that the weight gain and specific growth rate of fish in P2 and P0.5 groups were significantly higher than those in control P0 group at 30 and 60 days, respectively (P < 0.05). The antioxidant enzymes activity of catalase and superoxide dismutase in serum were significantly increased in P0.5/P1/P2 groups after 30 days. The transcriptional levels of penicillin-binding protein A and interleukin-8 analyzed by qRT-PCR were significantly upregulated in P2 and P4 groups compared to those in P0/P0.5/P1/P8 groups at 30 days. The transcriptional level of major histocompatibility complex class II in P2 group was significantly upregulated, and aldehyde oxidase downregulated compared to P0 group. Intestinal microbiota analysis by Illumina high-throughput sequencing showed that the microbiota diversity was not changed significantly, but the microbiota structure shifted significantly post PHB treatment. At the phyla level, Firmicutes and Proteobacteria were predominant in both P0 and P2 groups. At the genus level, the relative abundance of Bacillus spp. in P2 group increased significantly, and abundance of Achromobacter spp. decreased significantly. KEGG pathway analysis by PICRUSt showed that oral administration PHB significantly upregulated abundances of genes responsible for 10 pathways and downregulated genes involved in 17 pathways. In conclusion, soiny mullet fed with 2% PHB supplemental diets for 30 days showed better growth performance, higher antioxidant enzymes activity and immune-related genes expression. Their regulation of growth and immunity might be related with the intestinal microbiota change post PHB supplementation. It will provide very useful basic information to study the regulation mechanism of PHB in aquatic animals, and provide good green method to prevent disease in soiny mullet.


Subject(s)
Adjuvants, Immunologic/metabolism , Gastrointestinal Microbiome , Hydroxybutyrates/metabolism , Polyesters/metabolism , Smegmamorpha/immunology , Adjuvants, Immunologic/administration & dosage , Animal Feed/analysis , Animals , Diet/veterinary , Dietary Supplements/analysis , Gastrointestinal Microbiome/drug effects , Hydroxybutyrates/administration & dosage , Intestines/microbiology , Polyesters/administration & dosage , Smegmamorpha/growth & development , Smegmamorpha/microbiology
13.
Curr Microbiol ; 76(6): 755-761, 2019 Jun.
Article in English | MEDLINE | ID: mdl-29754180

ABSTRACT

Termites are global pests and can cause serious damage to buildings, crops, and plantation forests. The symbiotic intestinal flora plays an important role in the digestion of cellulose and nitrogen in the life of termites. Termites and their symbiotic microbes in the gut form a synergistic system. These organism work together to digest lignocellulose to make the termites grow on nitrogen deficient food. In this paper, the diversity of symbiotic microorganisms in the gut of termites, including protozoan, spirochetes, actinomycetes, fungus and bacteria, and their role in the digestion of lignocellulose and also the biotechnological applications of these symbiotic microorganisms are discussed. The high efficiency lignocellulose degradation systems of symbiotic microbes in termite gut not only provided a new way of biological energy development, but also has immense prospect in the application of cellulase enzymes. In addition, the study on the symbiotic microorganisms in the gut of termites will also provide a new method for the biological control of termites by the endophytic bacteria in the gut of termites.


Subject(s)
Bacteria/metabolism , Biodiversity , Biotechnology/methods , Fungi/metabolism , Isoptera/microbiology , Oxymonadida/metabolism , Parabasalidea/metabolism , Animals , Bacteria/classification , Bacteria/growth & development , Fungi/classification , Fungi/growth & development , Intestines/microbiology , Intestines/parasitology , Isoptera/parasitology , Lignin/metabolism , Oxymonadida/classification , Oxymonadida/growth & development , Parabasalidea/classification , Parabasalidea/growth & development , Symbiosis
14.
BMC Microbiol ; 18(1): 10, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29378521

ABSTRACT

BACKGROUND: Two-thirds of the world's population is thought to be infected by Helicobacter pylori. Although most people infected with H. pylori are asymptomatic, this pathogen is associated with several gastric pathologies including cancer. The risk factors for colonization are still unclear and the genetic diversity within individual hosts has never been clearly investigated. RESULT: This study determined the prevalence of, and explored risk factors for, H. pylori infection directly from paired saliva (n = 110) and stool (n = 110) samples from asymptomatic persons in Northeast Thailand. Samples were subjected to indirect immunofluorescence assay (IFA), 16S rRNA-based real-time PCR and vacA-based semi-nested PCR. Partial vacA gene sequences of H. pylori were compared between saliva and stool samples. The overall prevalence of H. pylori infection in our asymptomatic study population was 64%. Age, gender, occupation and frequency of brushing teeth were not found to be associated with H. pylori colonization. The vacA gene was successfully sequenced from both saliva and stool samples of 12 individuals. For seven of these individuals, saliva and stool sequences fell into different clusters on a phylogenetic tree, indicating intra-host genetic variation of H. pylori. CONCLUSION: This study reports a high prevalence of H. pylori infection in asymptomatic persons in this region of Thailand and demonstrates that genotypes (vacA gene sequences) of H. pylori may differ between the oral cavity and intestinal tract.


Subject(s)
Feces/microbiology , Genotype , Helicobacter Infections/epidemiology , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/isolation & purification , Saliva/microbiology , Adolescent , Adult , Bacterial Proteins/genetics , DNA, Bacterial , Female , Genetic Variation , Helicobacter pylori/classification , Helicobacter pylori/pathogenicity , Humans , Intestines/microbiology , Male , Middle Aged , Mouth/microbiology , Phylogeny , Polymerase Chain Reaction , Prevalence , RNA, Ribosomal, 16S/genetics , Risk Factors , Thailand/epidemiology , Young Adult
15.
Proc Natl Acad Sci U S A ; 112(35): 11060-5, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26283357

ABSTRACT

Despite the critical role of the human microbiota in health, our understanding of microbiota compositional dynamics during and after pregnancy is incomplete. We conducted a case-control study of 49 pregnant women, 15 of whom delivered preterm. From 40 of these women, we analyzed bacterial taxonomic composition of 3,767 specimens collected prospectively and weekly during gestation and monthly after delivery from the vagina, distal gut, saliva, and tooth/gum. Linear mixed-effects modeling, medoid-based clustering, and Markov chain modeling were used to analyze community temporal trends, community structure, and vaginal community state transitions. Microbiota community taxonomic composition and diversity remained remarkably stable at all four body sites during pregnancy (P > 0.05 for trends over time). Prevalence of a Lactobacillus-poor vaginal community state type (CST 4) was inversely correlated with gestational age at delivery (P = 0.0039). Risk for preterm birth was more pronounced for subjects with CST 4 accompanied by elevated Gardnerella or Ureaplasma abundances. This finding was validated with a set of 246 vaginal specimens from nine women (four of whom delivered preterm). Most women experienced a postdelivery disturbance in the vaginal community characterized by a decrease in Lactobacillus species and an increase in diverse anaerobes such as Peptoniphilus, Prevotella, and Anaerococcus species. This disturbance was unrelated to gestational age at delivery and persisted for up to 1 y. These findings have important implications for predicting premature labor, a major global health problem, and for understanding the potential impact of a persistent, altered postpartum microbiota on maternal health, including outcomes of pregnancies following short interpregnancy intervals.


Subject(s)
Microbiota , Female , Humans , Intestines/microbiology , Periodontium/microbiology , Pregnancy , Saliva/microbiology , Vagina/microbiology
16.
Microb Pathog ; 112: 5-14, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28942174

ABSTRACT

Inflammatory bowel disease (IBD) symbolizes a group of intestinal disorders in which prolonged inflammation occur in the digestive tract (esophagus, large intestine, small intestine mouth, stomach). Both genetic and environmental factors (infections, stress, diet) are involved in the development of IBD. As we know that bacteria are found in the intestinal mucosa of human and clinical observations revealed bacterial biofilms associated with patients of IBD. Various factors and microbes are found to play an essential role in biofilm formation and mucosal colonization during IBD. Biofilm formation in the digestive tract is dependent on an extracellular matrix synthesized by the bacteria and it has an adverse effect on the immune response of the host. There is no satisfactory and safe treatment option for IBD. Therefore, the current research aims to disrupt biofilm in IBD and concentrates predominantly on improving the drug. Here, we review the literature on bacterial biofilm and IBD to gather new knowledge on the current understanding of biofilm formation in IBD, host immune deregulation and dysbiosis in IBD, molecular mechanism, bacteria involved in biofilm formation, current and future regimen. It is urgently required to plan new ways to control and eradicate bacteria in biofilms that will open up novel diagnostic and therapeutic avenues for IBD. This article includes the mechanism of signaling molecules with respect to the biofilm-related genes as well as the diagnostic methods and new technologies involved in the treatment of IBD.


Subject(s)
Bacteria/immunology , Biofilms/growth & development , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Anti-Inflammatory Agents/pharmacology , Bacteria/drug effects , Bacteria/pathogenicity , Cyclic GMP/analogs & derivatives , Dysbiosis , Humans , Immunosuppressive Agents/pharmacology , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestines/immunology , Intestines/microbiology , Quorum Sensing , Stomach/microbiology
17.
Trop Med Int Health ; 22(6): 670-678, 2017 06.
Article in English | MEDLINE | ID: mdl-28319300

ABSTRACT

OBJECTIVE: To characterise childhood mouthing behaviours and to investigate the association between object-to-mouth and food-to-mouth contacts, diarrhoea prevalence and environmental enteropathy. METHODS: A prospective cohort study was conducted of 216 children ≤30 months of age in rural Bangladesh. Mouthing contacts with soil and food and objects with visible soil were assessed by 5-h structured observation. Stool was analysed for four faecal markers of intestinal inflammation: alpha-1-antitrypsin, myeloperoxidase, neopterin and calprotectin. RESULTS: Overall 82% of children were observed mouthing soil, objects with visible soil, or food with visible soil during the structured observation period. Sixty two percent of children were observed mouthing objects with visible soil, 63% were observed mouthing food with visible soil, and 18% were observed mouthing soil only. Children observed mouthing objects with visible soil had significantly elevated faecal calprotectin concentrations (206.81 µg/g, 95% confidence interval [CI]: 6.27, 407.36). There was also a marginally significant association between Escherichia coli counts in soil from a child's play space and the prevalence rate of diarrhoea (diarrhoea prevalence ratio: 2.03, 95% CI 0.97, 4.25). CONCLUSION: These findings provide further evidence to support the hypothesis that childhood mouthing behaviour in environments with faecal contamination can lead to environmental enteropathy in susceptible paediatric populations. Furthermore, these findings suggest that young children mouthing objects with soil, which occurred more frequently than soil directly (60% vs. 18%), was an important exposure route to faecal pathogens and a risk factor for environmental enteropathy.


Subject(s)
Child Behavior , Diarrhea/etiology , Environmental Exposure/adverse effects , Inflammation/etiology , Intestinal Diseases/etiology , Mouth , Soil , Bangladesh/epidemiology , Child, Preschool , Diarrhea/epidemiology , Diarrhea/microbiology , Escherichia coli , Feces/chemistry , Female , Humans , Infant , Inflammation/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/metabolism , Intestines/microbiology , Intestines/pathology , Leukocyte L1 Antigen Complex/metabolism , Male , Play and Playthings , Prospective Studies , Rural Population , Soil Microbiology
18.
Adv Appl Microbiol ; 98: 1-29, 2017.
Article in English | MEDLINE | ID: mdl-28189153

ABSTRACT

Probiotics are nonpathogenic microorganisms that confer a health benefit on the host when administered in adequate amounts. Ample evidence is documented to support the potential application of probiotics for the prevention and treatment of infections. Health benefits of probiotics include prevention of diarrhea, including antibiotic-associated diarrhea and traveler's diarrhea, atopic eczema, dental carries, colorectal cancers, and treatment of inflammatory bowel disease. The cumulative body of scientific evidence that demonstrates the beneficial effects of probiotics on health and disease prevention has made probiotics increasingly important as a part of human nutrition and led to a surge in the demand for probiotics in clinical applications and as functional foods. The ability of probiotics to promote health is attributed to the various beneficial effects exerted by these microorganisms on the host. These include lactose metabolism and food digestion, production of antimicrobial peptides and control of enteric infections, anticarcinogenic properties, immunologic enhancement, enhancement of short-chain fatty acid production, antiatherogenic and cholesterol-lowering attributes, regulatory role in allergy, protection against vaginal or urinary tract infections, increased nutritional value, maintenance of epithelial integrity and barrier, stimulation of repair mechanism in cells, and maintenance and reestablishment of well-balanced indigenous intestinal and respiratory microbial communities. Most of these attributes primarily focus on the effect of probiotic supplementation on the host. Hence, in most cases, it can be concluded that the ability of a probiotic to protect the host from infection is an indirect result of promoting overall health and well-being. However, probiotics also exert a direct effect on invading microorganisms. The direct modes of action resulting in the elimination of pathogens include inhibition of pathogen replication by producing antimicrobial substances like bacteriocins, competition for limiting resources in the host, antitoxin effect, inhibition of virulence, antiadhesive and antiinvasive effects, and competitive exclusion by competition for binding sites or stimulation of epithelial barrier function. Although much has been documented about the ability of probiotics to promote host health, there is limited discussion on the above mentioned effects of probiotics on pathogens. Being in an era of antibiotic resistance, a better understanding of this complex probiotic-pathogen interaction is critical for development of effective strategies to control infections. Therefore, this chapter will focus on the ability of probiotics to directly modulate the infectious nature of pathogens and the underlying mechanisms that mediate these effects.


Subject(s)
Drug Therapy , Gastrointestinal Microbiome , Lactobacillaceae/physiology , Probiotics/administration & dosage , Animals , Gastrointestinal Tract/microbiology , Humans , Intestines/microbiology , Lactobacillaceae/pathogenicity
19.
Int J Syst Evol Microbiol ; 67(9): 3363-3368, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28857023

ABSTRACT

Two strains (pika_113T and pika_114) of a previously undescribed Actinomyces-like bacterium were recovered from the intestinal contents of plateau pika (Ochotona curzoniae) on the Tibet-Qinghai Plateau, China. Results from biochemical characterization indicated that the two strains were phenotypically homogeneous and distinct from other previously described species of the genus Actinomyces. Based on the comparison of 16S rRNA gene sequences and genome analysis, the bacteria were determined to be a hitherto unknown subline within the genus Actinomyces, being most closely related to type strains of Actinomyces denticolens and Actinomyces timonensis with a respective 97.2 and 97.1 % similarity in their 16S rRNA gene sequences. Phylogenetic analyses confirmed that pika_113T was well separated from any other recognized species of the genus Actinomyces and within the cluster with A. denticolens and A. timonensis. The genome of strain pika_113T displayed less than 42 % relatedness in DNA-DNA hybridization with all the available genomes of existing species of the genus Actinomyces in the NCBI database. Collectively, based on the phenotypic characteristics and phylogenetic analyses results, we propose the novel isolates as representatives of Actinomyces gaoshouyii sp. nov. The type strain of Actinomyces gaoshouyii is pika_113T (=CGMCC 4.7372T=DSM 104049T), with a genomic DNA G+C content of 71 mol%.


Subject(s)
Actinomyces/classification , Intestines/microbiology , Lagomorpha/microbiology , Phylogeny , Actinomyces/genetics , Actinomyces/isolation & purification , Animals , Bacterial Typing Techniques , DNA, Bacterial/genetics , Nucleic Acid Hybridization , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Tibet
20.
Hepatology ; 62(4): 1260-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25820757

ABSTRACT

UNLABELLED: Altered gut microbiome is associated with systemic inflammation and cirrhosis decompensation. However, the correlation of the oral microbiome with inflammation in cirrhosis is unclear. Our aim was to evaluate the oral microbiome in cirrhosis and compare with stool microbiome. Outpatients with cirrhosis (with/without hepatic encephalopathy [HE]) and controls underwent stool/saliva microbiome analysis (for composition and function) and also systemic inflammatory evaluation. Ninety-day liver-related hospitalizations were recorded. Salivary inflammation was studied using T helper 1 cytokines/secretory immunoglobulin A (IgA), histatins and lysozyme in a subsequent group. A total of 102 patients with cirrhosis (43 previous HE) and 32 age-matched controls were included. On principal component analysis (PCA), stool and saliva microbiome clustered far apart, showing differences between sites as a whole. In salivary microbiome, with previous HE, relative abundance of autochthonous families decreased whereas potentially pathogenic ones (Enterobacteriaceae, Enterococcaceae) increased in saliva. Endotoxin-related predicted functions were significantly higher in cirrhotic saliva. In stool microbiome, relative autochthonous taxa abundance reduced in previous HE, along with increased Enterobacteriaceae and Enterococcaceae. Cirrhotic stool microbiota demonstrated a significantly higher correlation with systemic inflammation, compared to saliva microbiota, on correlation networks. Thirty-eight patients were hospitalized within 90 days. Their salivary dysbiosis was significantly worse and predicted this outcome independent of cirrhosis severity. Salivary inflammation was studied in an additional 86 age-matched subjects (43 controls/43 patients with cirrhosis); significantly higher interleukin (IL)-6/IL-1ß, secretory IgA, and lower lysozyme, and histatins 1 and 5 were found in patients with cirrhosis, compared to controls. CONCLUSIONS: Dysbiosis, represented by reduction in autochthonous bacteria, is present in both saliva and stool in patients with cirrhosis, compared to controls. Patients with cirrhosis have impaired salivary defenses and worse inflammation. Salivary dysbiosis was greater in patients with cirrhosis who developed 90-day hospitalizations. These findings could represent a global mucosal-immune interface change in cirrhosis.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome , Hepatic Encephalopathy/microbiology , Intestines/microbiology , Liver Cirrhosis/microbiology , Saliva/microbiology , Dysbiosis , Female , Hepatic Encephalopathy/complications , Humans , Inflammation/microbiology , Liver Cirrhosis/complications , Male , Microbiota , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL