626 results on '"intestinal dysbiosis"'
Search Results
602. Effects on intestinal microflora during systemic antimicrobial prophylaxis in orthopaedic patients: teicoplanin (TC) vs cefalozin (CZ)
- Author
-
BERTAZZONI MINELLI, Elisa, Benini, Anna, Barzoi, E., De Bastiani, G., and Periti, P.
- Subjects
antibacterial prophylaxis ,orthopedics ,Intesinal bacterial flora ,intestinal dysbiosis ,antibiotics - Published
- 1992
603. W2031 Antibiotic-Induced Intestinal Dysbiosis Results in Altered Behavior in BALB/C Mice
- Author
-
Wendy Jackson, Premysl Bercik, Greg Armstrong, Emmanuel Denou, Deng Yikang, Elena F. Verdu, Ailish Coblentz, Jennifer Jury, and Stephen M. Collins
- Subjects
Hepatology ,biology ,medicine.drug_class ,business.industry ,Antibiotics ,Gastroenterology ,medicine ,Intestinal dysbiosis ,biology.organism_classification ,business ,BALB/c ,Microbiology - Published
- 2009
- Full Text
- View/download PDF
604. Nutrition, oxidative stress and intestinal dysbiosis: Influence of diet on gut microbiota in inflammatory bowel diseases.
- Author
-
Tomasello G, Mazzola M, Leone A, Sinagra E, Zummo G, Farina F, Damiani P, Cappello F, Gerges Geagea A, Jurjus A, Bou Assi T, Messina M, and Carini F
- Subjects
- Dysbiosis diet therapy, Humans, Nutritional Status physiology, Oxidative Stress physiology, Diet, Dysbiosis etiology, Gastrointestinal Microbiome physiology, Inflammatory Bowel Diseases etiology
- Abstract
Background: Microbiota refers to the population of microorganisms (bacteria, viruses and fungi) that inhabit the entire gastrointestinal tract, more particularly the colon whose role is to maintain the integrity of the intestinal mucosa and control the proliferation of pathogenic bacteria. Alteration in the composition of the gut microbiota is called dysbiosis. Dysbiosis redisposes to inflammatory bowel diseases such as ulcerative colitis, Crohn disease and indeterminate colitis., Methods: The purpose of this literature review is to elucidate the influence of diet on the composition of the gastrointestinal microbiota in the healthy gut and the role of diet in the development of dysbiosis., Conclusion: The "Western diet", in particular a low - fiber high fat/high carbohydrate diet is one factor that can lead to severe dysbiosis. In contrast, "mediterranean" and vegetarian diets that includes abundant fruits, vegetables, olive oil and oily fish are known for their anti-inflammatory effects and could prevent dysbiosis and subsequent inflammatory bowel disease.
- Published
- 2016
- Full Text
- View/download PDF
605. Modulating the Gut Micro-Environment in the Treatment of Intestinal Parasites.
- Author
-
Vitetta L, Saltzman ET, Nikov T, Ibrahim I, and Hall S
- Abstract
The interactions of micro-organisms cohabitating with Homo sapiens spans millennia, with microbial communities living in a symbiotic relationship with the host. Interacting to regulate and maintain physiological functions and immunological tolerance, the microbial community is able to exert an influence on host health. An example of micro-organisms contributing to an intestinal disease state is exhibited by a biodiverse range of protozoan and bacterial species that damage the intestinal epithelia and are therefore implicated in the symptoms of diarrhea. As a contentious exemplar, Blastocystis hominis is a ubiquitous enteric protist that can adversely affect the intestines. The symptoms experienced are a consequence of the responses of the innate immune system triggered by the disruption of the intestinal barrier. The infiltration of the intestinal epithelial barrier involves a host of immune receptors, including toll like receptors and IgM/IgG/IgA antibodies as well as CD8+ T cells, macrophages, and neutrophils. Whilst the mechanisms of interactions between the intestinal microbiome and protozoan parasites remain incompletely understood, it is acknowledged that the intestinal microbiota is a key factor in the pathophysiology of parasitic infections. Modulating the intestinal environment through the administration of probiotics has been postulated as a possible therapeutic agent to control the proliferation of intestinal microbes through their capacity to induce competition for occupation of a common biotype. The ultimate goal of this mechanism is to prevent infections of the like of giardiasis and eliminate its symptoms. The differing types of probiotics (i.e., bacteria and yeast) modulate immunity by stimulating the host immune system. Early animal studies support the potential benefits of probiotic administration to prevent intestinal infections, with human clinical studies showing probiotics can reduce the number of parasites and the severity of symptoms. The early clinical indications endorse probiotics as adjuncts in the pharmaceutical treatment of protozoan infections. Currently, the bar is set low for the conduct of well-designed clinical studies that will translate the use of probiotics to ameliorate protozoan infections, therefore the requisite is for further clinical research., Competing Interests: Luis Vitetta has received National Institute of Complementary Medicine and National Health and Medical Research Council of Australia competitive funding and Industry support for research into probiotics.
- Published
- 2016
- Full Text
- View/download PDF
606. Altered gut microbiota in Rett syndrome.
- Author
-
Strati F, Cavalieri D, Albanese D, De Felice C, Donati C, Hayek J, Jousson O, Leoncini S, Pindo M, Renzi D, Rizzetto L, Stefanini I, Calabrò A, and De Filippo C
- Subjects
- Bacteria genetics, Biodiversity, Constipation pathology, Fatty Acids metabolism, Fungi genetics, Humans, Inflammation pathology, Metabolomics methods, Metagenomics methods, Methyl-CpG-Binding Protein 2 genetics, Rett Syndrome genetics, Bacteria classification, Dysbiosis microbiology, Fungi classification, Gastrointestinal Microbiome physiology, Intestines microbiology, Rett Syndrome microbiology, Rett Syndrome physiopathology
- Abstract
Background: The human gut microbiota directly affects human health, and its alteration can lead to gastrointestinal abnormalities and inflammation. Rett syndrome (RTT), a progressive neurological disorder mainly caused by mutations in MeCP2 gene, is commonly associated with gastrointestinal dysfunctions and constipation, suggesting a link between RTT's gastrointestinal abnormalities and the gut microbiota. The aim of this study was to evaluate the bacterial and fungal gut microbiota in a cohort of RTT subjects integrating clinical, metabolomics and metagenomics data to understand if changes in the gut microbiota of RTT subjects could be associated with gastrointestinal abnormalities and inflammatory status., Results: Our findings revealed the occurrence of an intestinal sub-inflammatory status in RTT subjects as measured by the elevated values of faecal calprotectin and erythrocyte sedimentation rate. We showed that, overall, RTT subjects harbour bacterial and fungal microbiota altered in terms of relative abundances from those of healthy controls, with a reduced microbial richness and dominated by microbial taxa belonging to Bifidobacterium, several Clostridia (among which Anaerostipes, Clostridium XIVa, Clostridium XIVb) as well as Erysipelotrichaceae, Actinomyces, Lactobacillus, Enterococcus, Eggerthella, Escherichia/Shigella and the fungal genus Candida. We further observed that alterations of the gut microbiota do not depend on the constipation status of RTT subjects and that this dysbiotic microbiota produced altered short chain fatty acids profiles., Conclusions: We demonstrated for the first time that RTT is associated with a dysbiosis of both the bacterial and fungal component of the gut microbiota, suggesting that impairments of MeCP2 functioning favour the establishment of a microbial community adapted to the costive gastrointestinal niche of RTT subjects. The altered production of short chain fatty acids associated with this microbiota might reinforce the constipation status of RTT subjects and contribute to RTT gastrointestinal physiopathology.
- Published
- 2016
- Full Text
- View/download PDF
607. The Role of Gut Microflora and the Cholinergic Anti-inflammatory Neuroendocrine System in Diabetes Mellitus.
- Author
-
Parekh PJ, Nayi VR, Johnson DA, and Vinik AI
- Abstract
The obesity epidemic has drastically impacted the state of health care in the United States. Paralleling this epidemic is the incidence of diabetes mellitus, with a notable shift toward a much younger age of onset. While central to the pathogenesis of diabetes associated with obesity is the role of inflammation attributed to "adiposopathy." Emerging data suggest that changes in sympathetic/parasympathetic balance regulated by the brain precede changes in the inflammatory cascade. It has now been established that the gut microflora contributes significantly to the activation and inhibition of autonomic control and impact the set of the neuroinflammatory inhibitory reflex mediated by the cholinergic nervous system. There has been a paradigm shift toward further investigating commensal bacteria in the pathogenesis of obesity and diabetes mellitus and its complications, as dysbiosis is thought to play a pivotal role in diabetic-associated disorders. This paper is intended to evaluate the role of intestinal dysbiosis in the pathogenesis of diabetes mellitus and examine the potential for restoration of balance via use of probiotics.
- Published
- 2016
- Full Text
- View/download PDF
608. Peripheral aetiopathogenic drivers and mediators of Parkinson's disease and co-morbidities: role of gastrointestinal microbiota.
- Author
-
Dobbs SM, Dobbs RJ, Weller C, Charlett A, Augustin A, Taylor D, Ibrahim MA, and Bjarnason I
- Subjects
- Animals, CD4-Positive T-Lymphocytes microbiology, CD4-Positive T-Lymphocytes pathology, Cognition Disorders epidemiology, Cognition Disorders pathology, Comorbidity, Depression epidemiology, Depression pathology, Dysbiosis epidemiology, Dysbiosis microbiology, Dysbiosis pathology, Helicobacter Infections epidemiology, Helicobacter Infections microbiology, Helicobacter Infections pathology, Helicobacter pylori growth & development, Helicobacter pylori pathogenicity, Humans, Inflammation, Inflammatory Bowel Diseases epidemiology, Inflammatory Bowel Diseases pathology, Killer Cells, Natural microbiology, Killer Cells, Natural pathology, Neutrophils microbiology, Neutrophils pathology, Parkinson Disease epidemiology, Parkinson Disease pathology, Peptic Ulcer epidemiology, Peptic Ulcer microbiology, Peptic Ulcer pathology, Cognition Disorders microbiology, Depression microbiology, Gastrointestinal Microbiome, Inflammatory Bowel Diseases microbiology, Parkinson Disease microbiology
- Abstract
We seek an aetiopathogenic model for the spectrum of Parkinson's disease (PD), functional bowel disease, depression and cognitive impairment. The adopted concept is that systemic immuno-inflammatory processes mediate neuro-inflammation. The model would be based on phenotype, exposome (including gastrointestinal microbiome), milieu (immuno-inflammatory and metabolome), human genetics and their interactions. It would enable a patient's position, to be understood in terms of drivers, perpetuators and mediators, and a future position, with and without intervention, predicted. Even the cardinal facets of PD may have different drivers: halting one may allow escape down subordinate pathways. Peptic ulceration is prodromal to PD. In our randomised placebo-controlled trial, hypokinesia improved over the year following biopsy-proven Helicobacter pylori eradication and rigidity worsened. This was independent of any (stable, long t½) antiparkinsonian medication. There are pointers to an autoimmune process: for example, surveillance-confirmed hypokinesia effect was indication specific. During surveillance, successive antimicrobial courses, other than for Helicobacter, were associated with cumulative increase in rigidity. Exhibiting laxatives appeared to stem the overall temporal increase, despite antiparkinsonian medication, in rigidity. Thus, intestinal dysbiosis may be a major source of bystander neuronal damage. There are biological gradients of objective measures of PD facets on circulating inflammatory markers and leucocyte subset counts. Moreover, lactulose hydrogen breath test positivity for small-intestinal bacterial overgrowth (present in two thirds of PD patients) is associated with the same subsets: higher natural killer and total CD4+ counts and lower neutrophils. With greater aetiopathogenic understanding, relatively low cost and on-the-shelf medication could have a major impact. A new generation of animal models, based on the gut microbiome, is envisaged.
- Published
- 2016
- Full Text
- View/download PDF
609. Sleep quality and the treatment of intestinal microbiota imbalance in Chronic Fatigue Syndrome: A pilot study.
- Author
-
Jackson ML, Butt H, Ball M, Lewis DP, and Bruck D
- Abstract
Chronic Fatigue Syndrome (CFS) is a multisystem illness, which may be associated with imbalances in gut microbiota. This study builds on recent evidence that sleep may be influenced by gut microbiota, by assessing whether changes to microbiota in a clinical population known to have both poor sleep and high rates of colonization with gram-positive faecal Streptococcus, can improve sleep. Twenty-one CFS participants completed a 22- day open label trial. Faecal microbiota analysis was performed at baseline and at the end of the trial. Participants were administered erythromycin 400 mg b.d. for 6 days. Actigraphy and questionnaires were used to monitor sleep, symptoms and mood. Changes in patients who showed a clinically significant change in faecal Streptococcus after treatment (responders; defined as post-therapy distribution<6%) were compared to participants who did not respond to treatment. In the seven responders, there was a significant increase in actigraphic total sleep time (p=0.028) from baseline to follow up, compared with non-responders. Improved vigour scores were associated with a lower Streptococcus count (ρ=-0.90, p=0.037). For both the responders and the whole group, poorer mood was associated with higher Lactobacillus. Short term antibiotic treatment appears to be insufficient to effect sustainable changes in the gut ecosystem in most CFS participants. Some improvement in objective sleep parameters and mood were found in participants with reduced levels of gram-positive gut microbiota after antibiotic treatment, which is encouraging. Further study of possible links between gut microorganisms and sleep and mood disturbances is warranted.
- Published
- 2015
- Full Text
- View/download PDF
610. Fecal microbiota transplantation and bacterial consortium transplantation have comparable effects on the re-establishment of mucosal barrier function in mice with intestinal dysbiosis.
- Author
-
Li M, Liang P, Li Z, Wang Y, Zhang G, Gao H, Wen S, and Tang L
- Abstract
Fecal microbiota transplantation (FMT) is a promising therapy, despite some reports of adverse side effects. Bacterial consortia transplantation (BCT) for targeted restoration of the intestinal ecosystem is considered a relatively safe and simple procedure. However, no systematic research has assessed the effects of FMT and BCT on immune responses of intestinal mucosal barrier in patients. We conducted complementary studies in animal models on the effects of FMT and BCT, and provide recommendations for improving the clinical outcomes of these treatments. To establish the dysbiosis model, male BALB/c mice were treated with ceftriaxone intra-gastrically for 7 days. After that, FMT and BCT were performed on ceftriaxone-treated mice for 3 consecutive days to rebuild the intestinal ecosystem. Post-FMT and post-BCT changes of the intestinal microbial community and mucosal barrier functions were investigated and compared. Disruption of intestinal microbial homeostasis impacted the integrity of mucosal epithelial layer, resulting in increased intestinal permeability. These outcomes were accompanied by overexpression of Muc2, significant decrease of SIgA secretion, and overproduction of defensins and inflammatory cytokines. After FMT and BCT, the intestinal microbiota recovered quickly, this was associated with better reconstruction of mucosal barriers and re-establishment of immune networks compared with spontaneous recovery (SR). Although based on a short-term study, our results suggest that FMT and BCT promote the re-establishment of intestinal microbial communities in mice with antibiotic-induced dysbiosis, and contribute to the temporal and spatial interactions between microbiota and mucosal barriers. The effects of BCT are comparable to that of FMT, especially in normalizing the intestinal levels of Muc2, SIgA, and defensins.
- Published
- 2015
- Full Text
- View/download PDF
611. The natural approach to osteoporosis.
- Author
-
Bartolozzi E
- Abstract
Osteoporosis is normally the result of a wrong life-style (diet, physical inactivity, smoke, dental hygiene, intestinal dysbiosis,…) and environmental toxicity which stimulate the chronic expression of inflammatory genes and alter the immuno-endocrine balance. A natural approch should face all the factors involved, leading the patients to become aware of their own responsability, and helping them with natural therapies, healthy food and life-style which support their body in the process of self-healing.
- Published
- 2015
- Full Text
- View/download PDF
612. Potential use of probiotic and commensal bacteria as non-antibiotic strategies against vancomycin-resistant enterococci.
- Author
-
Crouzet L, Rigottier-Gois L, and Serror P
- Subjects
- Animals, Anti-Bacterial Agents adverse effects, Disease Models, Animal, Humans, Vancomycin-Resistant Enterococci pathogenicity, Bacteria growth & development, Gastrointestinal Tract microbiology, Probiotics administration & dosage, Symbiosis, Vancomycin-Resistant Enterococci growth & development
- Abstract
Vancomycin-resistant enterococci (VRE) represent major opportunistic pathogens in immunocompromised populations. Particularly adapted to the hospital environment, VRE efficiently colonize the gastrointestinal (GI) tract of patients. Furthermore, certain circumstances of antibiotic-induced dysbiosis of the gut microbiota contribute to colonization, overgrowth and persistence of VRE in the GI tract, increasing the risk of infection in critically ill and/or severally immunocompromised patients. VRE treatment with antibiotics remains challenging due to the robustness and ability of enterococci to adapt to harsh conditions and to acquire novel resistance genes. Reducing VRE intestinal colonization, overgrowth and carriage has thus become an important issue to reduce the risk of infection and dissemination. This review summarizes the knowledge of the conditions favoring VRE colonization and persistence in the GI tract and focuses on the strategies to reduce overgrowth or persistence of VRE in the GI tract based on the oral administration of probiotic or commensal bacteria in human studies and in animal models, and on the potential underlying mechanisms of the anti-VRE effect., (© FEMS 2015. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
613. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice.
- Author
-
Aguilera M, Cerdà-Cuéllar M, and Martínez V
- Subjects
- Animals, Anti-Bacterial Agents administration & dosage, Colon drug effects, Colon pathology, Female, Mice, Anti-Bacterial Agents adverse effects, Colon microbiology, Colon physiology, Dysbiosis chemically induced, Gastrointestinal Microbiome drug effects
- Abstract
Alterations in the composition of the commensal microbiota (dysbiosis) seem to be a pathogenic component of functional gastrointestinal disorders, mainly irritable bowel syndrome (IBS), and might participate in the secretomotor and sensory alterations observed in these patients.We determined if a state antibiotics-induced intestinal dysbiosis is able to modify colonic pain-related and motor responses and characterized the neuro-immune mechanisms implicated in mice. A 2-week antibiotics treatment induced a colonic dysbiosis (increments in Bacteroides spp, Clostridium coccoides and Lactobacillus spp and reduction in Bifidobacterium spp). Bacterial adherence was not affected. Dysbiosis was associated with increased levels of secretory-IgA, up-regulation of the antimicrobial lectin RegIIIγ, and toll-like receptors (TLR) 4 and 7 and down-regulation of the antimicrobial-peptide Resistin-Like Molecule-β and TLR5. Dysbiotic mice showed less goblet cells, without changes in the thickness of the mucus layer. Neither macroscopical nor microscopical signs of inflammation were observed. In dysbiotic mice, expression of the cannabinoid receptor 2 was up-regulated, while the cannabinoid 1 and the mu-opioid receptors were down-regulated. In antibiotic-treated mice, visceral pain-related responses elicited by intraperitoneal acetic acid or intracolonic capsaicin were significantly attenuated. Colonic contractility was enhanced during dysbiosis. Intestinal dysbiosis induce changes in the innate intestinal immune system and modulate the expression of pain-related sensory systems, an effect associated with a reduction in visceral pain-related responses. Commensal microbiota modulates gut neuro-immune sensory systems, leading to functional changes, at least as it relates to viscerosensitivity. Similar mechanisms might explain the beneficial effects of antibiotics or certain probiotics in the treatment of IBS.
- Published
- 2015
- Full Text
- View/download PDF
614. The intestinal microbiome in early life: health and disease.
- Author
-
Arrieta MC, Stiemsma LT, Amenyogbe N, Brown EM, and Finlay B
- Abstract
Human microbial colonization begins at birth and continues to develop and modulate in species abundance for about 3 years, until the microbiota becomes adult-like. During the same time period, children experience significant developmental changes that influence their health status as well as their immune system. An ever-expanding number of articles associate several diseases with early-life imbalances of the gut microbiota, also referred to as gut microbial dysbiosis. Whether early-life dysbiosis precedes and plays a role in disease pathogenesis, or simply originates from the disease process itself is a question that is beginning to be answered in a few diseases, including IBD, obesity, and asthma. This review describes the gut microbiome structure and function during the formative first years of life, as well as the environmental factors that determine its composition. It also aims to discuss the recent advances in understanding the role of the early-life gut microbiota in the development of immune-mediated, metabolic, and neurological diseases. A greater understanding of how the early-life gut microbiota impacts our immune development could potentially lead to novel microbial-derived therapies that target disease prevention at an early age.
- Published
- 2014
- Full Text
- View/download PDF
615. Adult bile acid amino transferase deficiency.
- Author
-
Lord RS, Tuttle DM, and Cantor DS
- Abstract
Patient: Female, 70 FINAL DIAGNOSIS: Bile acid amino transferase deficiency Symptoms: Headache • indigestion • itching skin • nausea • vomiting, Medication: - Clinical Procedure: - Specialty: Gastroenterology and Hepatology., Objective: Challenging differential diagnosis., Background: Bile acid synthesis impairments are difficult to diagnose due to non-specific manifestations related to progressive failure to absorb essential fatty acids and fat soluble vitamins and failure to maintain normal intestinal microbiota., Case Report: A 70-year-old female presented with long-standing history of recurrent headaches, indigestion, dry, scaly, itching skin, and fluid around knee joints. Quantitative Electroencephalography (QEEG) revealed widespread excess theta maximum in the temporal regions. A rare pattern of elevated plasma glycine and taurine led to suspicion of BAATD. A stool profile employing molecular probes for commensal bacteria revealed elevation of Fusobacteria spp. Implementation of bile acid replacement therapy (BART) produced rapid remission of headache and other symptoms and a three-month follow up stool profile revealed normalization of fecal Fusobacteria populations that remained normal after one year of BART. QEEG analyses 4 weeks following BART showed evidence of significant improvement in CNS functioning., Conclusions: This case illustrates the potential for diagnosis of latent, adult BAATD by finding a unique pattern of plasma amino acids and monitoring of therapy by observing normalization of fecal commensal bacteria and functional brain assessments.
- Published
- 2014
- Full Text
- View/download PDF
616. Drosophila as a model for intestinal dysbiosis and chronic inflammatory diseases.
- Author
-
Lee KA and Lee WJ
- Subjects
- Animals, Chronic Disease, Dysbiosis microbiology, Genotype, Homeostasis, Host-Pathogen Interactions, Humans, Immunity genetics, Inflammation microbiology, Intestinal Diseases microbiology, Intestines microbiology, Microbiota, Models, Animal, Drosophila melanogaster immunology, Dysbiosis immunology, Inflammation immunology, Intestinal Diseases immunology, Intestines immunology
- Abstract
The association between deregulated intestinal microbial consortia and host diseases has been recognized since the birth of microbiology over a century ago. Intestinal dysbiosis refers to a state where living metazoans harbor harmful intestinal microflora. However, there is still an issue of whether causality arises from the host or the microbe because it is unclear whether deregulation of the gut microbiota community is the consequence or cause of the host disease. Recent studies using Drosophila and its simple microbiota have provided a valuable model system for dissecting the molecular mechanisms of intestinal dysbiosis. In this review, we examine recent exciting observations in Drosophila gut-microbiota interactions, particularly the links among the host immune genotype, the microbial community structure, and the host inflammatory phenotype. Future genetic analyses using Drosophila model system will provide a valuable outcome for understanding the evolutionarily conserved mechanisms that underlie intestinal dysbiosis and chronic inflammatory diseases., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
617. Stress and antibiotics alter luminal and wall-adhered microbiota and enhance the local expression of visceral sensory-related systems in mice.
- Author
-
Aguilera M, Vergara P, and Martínez V
- Subjects
- Animals, Colon metabolism, Female, Intestinal Mucosa metabolism, Mice, Mice, Inbred C57BL, Microbiota physiology, Random Allocation, Stress, Psychological metabolism, Up-Regulation physiology, Visceral Pain etiology, Visceral Pain metabolism, Anti-Bacterial Agents toxicity, Colon drug effects, Intestinal Mucosa drug effects, Microbiota drug effects, Stress, Psychological complications, Visceral Pain chemically induced
- Abstract
Background: Stress leads to altered gastrointestinal neuro-immune responses. We characterized the interaction between stress and gut commensal microbiota and their role modulating colonic responses to stress, the induction of inflammation, the expression of sensory-related markers, and visceral sensitivity., Methods: C57BL/6N female mice were treated (7 days, PO) with non-absorbable-broad spectrum antibiotics (bacitracin/neomycin, 0.4 mg per mouse per day). Simultaneously, mice were subjected to a 1 h per day (7 days) session of psychological stress (water avoidance stress, WAS). Luminal and wall-adhered microbiota were characterized by fluorescent in situ hybridization. Cannabinoid receptors 1 and 2 (CB1/2), tryptophan hydroxylase 1 and 2 (TPH1/2), and inflammatory markers were quantified by reverse transcription-quantitative real-time PCR (RT-qPCR) and secretory-IgA (s-IgA) by ELISA. Visceral sensitivity was assessed after the intracolonic administration of capsaicin., Key Results: Antibiotics did not affect the defecatory and endocrine responses to stress. However, antibiotics diminished by 2.5-folds total bacterial counts, induced a specific dysbiosis and favored bacterial wall adherence. Combining antibiotics and stress resulted in further reductions in bacterial counts and a dysbiosis, with enhanced bacterial wall adherence. Luminal s-IgA levels increased in dysbiotic mice. Nevertheless, no alterations consistent with the induction of colonic inflammation were observed. Dysbiosis upregulated CB2 expression and stress upregulated CB2 and TPH1 expression. Stress enhanced visceral pain-related responses, an effect prevented by antibiotic treatment., Conclusions & Inferences: Manipulations of the commensal microbiota and the interaction host-microbiota are able to modulate the local expression of neuro-immune-endocrine systems within the colon, leading to a modulation of visceral sensitivity. These mechanisms might contribute to the pathogenic and protective roles of microbiota in gastrointestinal homeostasis., (© 2013 John Wiley & Sons Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
618. Sleep quality and the treatment of intestinal microbiota imbalance in Chronic Fatigue Syndrome: A pilot study
- Author
-
Melinda L. Jackson, Michelle Ball, Dorothy Bruck, Henry L. Butt, and Donald P. Lewis
- Subjects
Chronic Fatigue Syndrome ,medicine.medical_specialty ,HPA, hypothalamic-pituitary adrenal ,lcsh:BF1-990 ,Population ,Neuroscience (miscellaneous) ,Medicine (miscellaneous) ,lcsh:Consciousness. Cognition ,Gut flora ,SFI, sleep fragmentation index ,CNS, central nervous system ,Behavioral Neuroscience ,Internal medicine ,Lactobacillus ,Full Length Article ,Mood ,Chronic fatigue syndrome ,medicine ,Psychiatry ,education ,TST, total sleep time ,POMS, Profile of Mood States ,education.field_of_study ,biology ,business.industry ,Actigraphy ,lcsh:BF309-499 ,biology.organism_classification ,medicine.disease ,Sleep in non-human animals ,3. Good health ,Clinical trial ,lcsh:Psychology ,FMA, faecal microbiota analysis ,CFS, Chronic Fatigue Syndrome ,Intestinal dysbiosis ,MALDI-TOF MS, matrix assisted laser desorption ionization – time of flight mass spectrometry ,WASO, wake after sleep onset ,SOL, sleep onset latency ,business ,Sleep - Abstract
Chronic Fatigue Syndrome (CFS) is a multisystem illness, which may be associated with imbalances in gut microbiota. This study builds on recent evidence that sleep may be influenced by gut microbiota, by assessing whether changes to microbiota in a clinical population known to have both poor sleep and high rates of colonization with gram-positive faecal Streptococcus, can improve sleep. Twenty-one CFS participants completed a 22-day open label trial. Faecal microbiota analysis was performed at baseline and at the end of the trial. Participants were administered erythromycin 400mg b.d. for 6 days. Actigraphy and questionnaires were used to monitor sleep, symptoms and mood. Changes in patients who showed a clinically significant change in faecal Streptococcus after treatment (responders; defined as post-therapy distribution
- Full Text
- View/download PDF
619. Peripheral aetiopathogenic drivers and mediators of Parkinson’s disease and co-morbidities: role of gastrointestinal microbiota
- Author
-
Dobbs, Sylvia M., Dobbs, R. John, Weller, Clive, Charlett, Andre, Augustin, Aisha Dominique, Taylor, David Michael, Ibrahim, Mohammad A A, and Bjarnason, Ingvar Iain
- Subjects
Cellular and Molecular Neuroscience ,Neurology ,Helicobacter ,Virology ,Intestinal dysbiosis ,Clinical Neurology ,Autoimmunity ,Pathogenesis ,Aetiology ,Bystander damage ,Parkinson’s and overlap diseases - Abstract
We seek an aetiopathogenic model for the spectrum of Parkinson’s disease (PD), functional bowel disease, depression and cognitive impairment. The adopted concept is that systemic immuno-inflammatory processes mediate neuro-inflammation. The model would be based on phenotype, exposome (including gastrointestinal microbiome), milieu (immuno-inflammatory and metabolome), human genetics and their interactions. It would enable a patient’s position, to be understood in terms of drivers, perpetuators and mediators, and a future position, with and without intervention, predicted. Even the cardinal facets of PD may have different drivers: halting one may allow escape down subordinate pathways. Peptic ulceration is prodromal to PD. In our randomised placebo-controlled trial, hypokinesia improved over the year following biopsy-proven Helicobacter pylori eradication and rigidity worsened. This was independent of any (stable, long t½) antiparkinsonian medication. There are pointers to an autoimmune process: for example, surveillance-confirmed hypokinesia effect was indication specific. During surveillance, successive antimicrobial courses, other than for Helicobacter, were associated with cumulative increase in rigidity. Exhibiting laxatives appeared to stem the overall temporal increase, despite antiparkinsonian medication, in rigidity. Thus, intestinal dysbiosis may be a major source of bystander neuronal damage. There are biological gradients of objective measures of PD facets on circulating inflammatory markers and leucocyte subset counts. Moreover, lactulose hydrogen breath test positivity for small-intestinal bacterial overgrowth (present in two thirds of PD patients) is associated with the same subsets: higher natural killer and total CD4+ counts and lower neutrophils. With greater aetiopathogenic understanding, relatively low cost and on-the-shelf medication could have a major impact. A new generation of animal models, based on the gut microbiome, is envisaged.
- Full Text
- View/download PDF
620. PREDICTORS OF DISORDER OF INTESTINAL MICROBIOCENOSIS OF THE FIRST-YEAR CHILDREN AND ITS PROPHYLAXIS
- Author
-
Kazakova, L. M., Slipachuk, L. V., Vasiukova, M. M., Stroi, O. A., Kazakova, L. M., Slipachuk, L. V., Vasiukova, M. M., and Stroi, O. A.
621. PECULIARITIES OF THE STATE OF THE INTESTINAL MICROBIOTA IN WOMEN WITH HABITUAL MISCARRIAGE
- Author
-
Pavlovska, O. M., Pavlovska, K. M., Krasnova, Zh. O., Pavlovska, O. M., Pavlovska, K. M., and Krasnova, Zh. O.
622. EFFICIENCY OF PATHOGENETIC THERAPY OF PATIENTS WITH ISCHEMIC HEART DISEASE WITH INTESTINAL DYSBIOSIS AND NORMOBIOTSENOZOM
- Author
-
Khimion, L. V., Busyhina, O. S., Produsevych, L. V., Khimion, L. V., Busyhina, O. S., and Produsevych, L. V.
623. Altered gut microbiota in Rett syndrome
- Author
-
Francesco Strati, Irene Stefanini, Silvia Leoncini, Lisa Rizzetto, Carlotta De Filippo, Claudio De Felice, Olivier Jousson, Daniela Renzi, Duccio Cavalieri, Claudio Donati, Antonino Salvatore Calabrò, Joussef Hayek, Davide Albanese, Massimo Pindo, Strati, F, Cavalieri, D, Albanese, D, De Felice, C, Donati, C, Hayek, J, Jousson, O, Leoncini, S, Pindo, M, Renzi, D, Rizzetto, L, Stefanini, I, Calabro, A, De Filippo, C, and Calabrò, A
- Subjects
SCFAs ,0301 basic medicine ,Methyl-CpG-Binding Protein 2 ,gut microbiome ,Intestinal dysbiosi ,Gut flora ,0302 clinical medicine ,Rett syndrome ,fluids and secretions ,Anaerostipes ,Mycobiota ,Lactobacillus ,Bifidobacterium ,2. Zero hunger ,biology ,Fatty Acids ,Gastrointestinal Microbiome ,Biodiversity ,3. Good health ,Intestines ,Intestinal dysbiosis ,Settore BIO/19 - MICROBIOLOGIA GENERALE ,Microbiology (medical) ,congenital, hereditary, and neonatal diseases and abnormalities ,food.ingredient ,Metataxonomic ,Gut microbiota ,Microbiology ,digestive system ,03 medical and health sciences ,food ,medicine ,Humans ,Metabolomics ,Inflammation ,Bacteria ,Research ,Metataxonomics ,Fungi ,SCFA ,biology.organism_classification ,medicine.disease ,030104 developmental biology ,Immunology ,Constipation ,Dysbiosis ,Metagenomics ,mycobiota ,030217 neurology & neurosurgery ,Actinomyces - Abstract
Background The human gut microbiota directly affects human health, and its alteration can lead to gastrointestinal abnormalities and inflammation. Rett syndrome (RTT), a progressive neurological disorder mainly caused by mutations in MeCP2 gene, is commonly associated with gastrointestinal dysfunctions and constipation, suggesting a link between RTT’s gastrointestinal abnormalities and the gut microbiota. The aim of this study was to evaluate the bacterial and fungal gut microbiota in a cohort of RTT subjects integrating clinical, metabolomics and metagenomics data to understand if changes in the gut microbiota of RTT subjects could be associated with gastrointestinal abnormalities and inflammatory status. Results Our findings revealed the occurrence of an intestinal sub-inflammatory status in RTT subjects as measured by the elevated values of faecal calprotectin and erythrocyte sedimentation rate. We showed that, overall, RTT subjects harbour bacterial and fungal microbiota altered in terms of relative abundances from those of healthy controls, with a reduced microbial richness and dominated by microbial taxa belonging to Bifidobacterium, several Clostridia (among which Anaerostipes, Clostridium XIVa, Clostridium XIVb) as well as Erysipelotrichaceae, Actinomyces, Lactobacillus, Enterococcus, Eggerthella, Escherichia/Shigella and the fungal genus Candida. We further observed that alterations of the gut microbiota do not depend on the constipation status of RTT subjects and that this dysbiotic microbiota produced altered short chain fatty acids profiles. Conclusions We demonstrated for the first time that RTT is associated with a dysbiosis of both the bacterial and fungal component of the gut microbiota, suggesting that impairments of MeCP2 functioning favour the establishment of a microbial community adapted to the costive gastrointestinal niche of RTT subjects. The altered production of short chain fatty acids associated with this microbiota might reinforce the constipation status of RTT subjects and contribute to RTT gastrointestinal physiopathology. Electronic supplementary material The online version of this article (doi:10.1186/s40168-016-0185-y) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
624. [Untitled]
- Subjects
0301 basic medicine ,Microbiology (medical) ,medicine.medical_specialty ,biology ,Firmicutes ,Bacteroidetes ,Intestinal dysbiosis ,Disease ,biology.organism_classification ,medicine.disease ,Microbiology ,Gastroenterology ,digestive system diseases ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Medical microbiology ,030225 pediatrics ,Meta-analysis ,Internal medicine ,Immunology ,Necrotizing enterocolitis ,medicine ,Microbiome - Abstract
Necrotizing enterocolitis (NEC) is a catastrophic disease of preterm infants, and microbial dysbiosis has been implicated in its pathogenesis. Studies evaluating the microbiome in NEC and preterm infants lack power and have reported inconsistent results. Our objectives were to perform a systematic review and meta-analyses of stool microbiome profiles in preterm infants to discern and describe microbial dysbiosis prior to the onset of NEC and to explore heterogeneity among studies. We searched MEDLINE, PubMed, CINAHL, and conference abstracts from the proceedings of Pediatric Academic Societies and reference lists of relevant identified articles in April 2016. Studies comparing the intestinal microbiome in preterm infants who developed NEC to those of controls, using culture-independent molecular techniques and reported α and β-diversity metrics, and microbial profiles were included. In addition, 16S ribosomal ribonucleic acid (rRNA) sequence data with clinical meta-data were requested from the authors of included studies or searched in public data repositories. We reprocessed the 16S rRNA sequence data through a uniform analysis pipeline, which were then synthesized by meta-analysis. We included 14 studies in this review, and data from eight studies were available for quantitative synthesis (106 NEC cases, 278 controls, 2944 samples). The age of NEC onset was at a mean ± SD of 30.1 ± 2.4 weeks post-conception (n = 61). Fecal microbiome from preterm infants with NEC had increased relative abundances of Proteobacteria and decreased relative abundances of Firmicutes and Bacteroidetes prior to NEC onset. Alpha- or beta-diversity indices in preterm infants with NEC were not consistently different from controls, but we found differences in taxonomic profiles related to antibiotic exposure, formula feeding, and mode of delivery. Exploring heterogeneity revealed differences in microbial profiles by study and the target region of the 16S rRNA gene (V1-V3 or V3-V5). Microbial dysbiosis preceding NEC in preterm infants is characterized by increased relative abundances of Proteobacteria and decreased relative abundances of Firmicutes and Bacteroidetes. Microbiome optimization may provide a novel strategy for preventing NEC.
625. The level of immunoglobulins in coprofiltrates and blood serum of children with pneumonia complicated by intestinal dysbiosis
- Author
-
A. V. Mitrofanova, L. L. Ksenofontova, L. I. Shchetinina, G. A. Katkova, T. V. Vertyashkina, T. N. Novoseltseva, L. V. Kurashvili, L. P. Remizova, and G. K. Bulanova
- Subjects
Pneumonia ,Blood serum ,biology ,business.industry ,Immunology ,biology.protein ,Medicine ,Intestinal dysbiosis ,General Medicine ,Acute pneumonia ,Antibody ,business ,medicine.disease - Abstract
The aim of this work was to study the content of immunoglobulins in blood serum and coprofiltrates in children with acute pneumonia, occurring against the background of intestinal dysbiosis.
- Published
- 1985
- Full Text
- View/download PDF
626. The incidence of intestinal dysbiosis in children
- Author
-
M. I. Zryachkin
- Subjects
medicine.medical_specialty ,business.industry ,Internal medicine ,Incidence (epidemiology) ,medicine ,Intestinal dysbiosis ,General Medicine ,business ,Gastroenterology - Abstract
Examination of 145 healthy children aged 1 month to 2 years was carried out. In 101 (69 6%) children, intestinal dysbiosis of varying severity was revealed. The frequency of detection and the degree of its severity depend on the age, type of feeding, the period since the last illness, and the number of antibacterial drugs taken earlier.
- Published
- 1982
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.