Articles de revues sur le sujet « Microbiota intestinale (Gut microbiota) »

Pour voir les autres types de publications sur ce sujet consultez le lien suivant : Microbiota intestinale (Gut microbiota).

Créez une référence correcte selon les styles APA, MLA, Chicago, Harvard et plusieurs autres

Choisissez une source :

Consultez les 50 meilleurs articles de revues pour votre recherche sur le sujet « Microbiota intestinale (Gut microbiota) ».

À côté de chaque source dans la liste de références il y a un bouton « Ajouter à la bibliographie ». Cliquez sur ce bouton, et nous générerons automatiquement la référence bibliographique pour la source choisie selon votre style de citation préféré : APA, MLA, Harvard, Vancouver, Chicago, etc.

Vous pouvez aussi télécharger le texte intégral de la publication scolaire au format pdf et consulter son résumé en ligne lorsque ces informations sont inclues dans les métadonnées.

Parcourez les articles de revues sur diverses disciplines et organisez correctement votre bibliographie.

1

Liu, Qihong, Yunfeng Luo et Xiao Ke. « Interaction between the Gut Microbiota and Intestinal Motility ». Evidence-Based Complementary and Alternative Medicine 2022 (15 novembre 2022) : 1–5. http://dx.doi.org/10.1155/2022/3240573.

Texte intégral
Résumé :
The gut microbiota is the largest symbiotic ecosystem with the host and has been proven to play an important role in maintaining the stability of the intestinal environment. The imbalance of the gut microbiota is caused by the imbalance between the symbiotic microbiota and the pathogenic microbiota. The commensal microbiome regulates intestinal motility, while the pathogenic microbiome causes intestinal motility disorder, resulting in disease development. Intestinal motility is a relatively general term, and its meaning may include intestinal muscle contraction, intestinal wall biomechanics, intestinal compliance, and transmission. The role of intestinal microecology and intestinal motility are interrelated, intestinal flora disorder mediates intestinal motility, and abnormal intestinal motility affects colonization of the intestinal flora. In this review, we briefly outlined the interaction between gut microbiota and intestinal motility and provided a reference for future studies.
Styles APA, Harvard, Vancouver, ISO, etc.
2

Thomas, Linda V., Theo Ockhuizen et Kaori Suzuki. « Exploring the influence of the gut microbiota and probiotics on health : a symposium report ». British Journal of Nutrition 112, S1 (23 juin 2014) : S1—S18. http://dx.doi.org/10.1017/s0007114514001275.

Texte intégral
Résumé :
The present report describes the presentations delivered at the 7th International Yakult Symposium, ‘The Intestinal Microbiota and Probiotics: Exploiting Their Influence on Health’, in London on 22–23 April 2013. The following two themes associated with health risks were covered: (1) the impact of age and diet on the gut microbiota and (2) the gut microbiota's interaction with the host. The strong influence of the maternal gut microbiota on neonatal colonisation was reported, as well as rapid changes in the gut microbiome of older people who move from community living to residential care. The effects of dietary changes on gut metabolism were described and the potential influence of inter-individual microbiota differences was noted, in particular the presence/absence of keystone species involved in butyrate metabolism. Several speakers highlighted the association between certain metabolic disorders and imbalanced or less diverse microbiota. Data from metagenomic analyses and novel techniques (including anex vivohuman mucosa model) provided new insights into the microbiota's influence on coeliac, obesity-related and inflammatory diseases, as well as the potential of probiotics.Akkermansia muciniphilaandFaecalibacterium prausnitziiwere suggested as targets for intervention. Host–microbiota interactions were explored in the context of gut barrier function, pathogenic bacteria recognition, and the ability of the immune system to induce either tolerogenic or inflammatory responses. There was speculation that the gut microbiota should be considered a separate organ, and whether analysis of an individual's microbiota could be useful in identifying their disease risk and/or therapy; however, more research is needed into specific diseases, different population groups and microbial interventions including probiotics.
Styles APA, Harvard, Vancouver, ISO, etc.
3

Meiliana, Anna, et Andi Wijaya. « Gut Microbiota, Obesity and Metabolic Dysfunction ». Indonesian Biomedical Journal 3, no 3 (1 décembre 2011) : 150. http://dx.doi.org/10.18585/inabj.v3i3.147.

Texte intégral
Résumé :
BACKGROUND: The prevalence of obesity and related disorders such as metabolic syndrome and diabetes has vastly increased throughout the world. Recent insights have generated an entirely new perspective suggesting that our microbiota might be involved in the development of these disorders. This represents an area of scientific need, opportunity and challenge. The insights gleaned should help to address several pressing global health problems.CONTENT: Our bowels have two major roles: the digestion and absorption of nutrients and the maintenance of a barrier against the external environment. They fulfill these functions in the context of, and with the help from, tens of trillions of resident microbes, known as the gut microbiota. Studies have demonstrated that obesity and metabolic syndrome may be associated with profound microbiotal changes, and the induction of a metabolic syndrome phenotype through fecal transplants corroborates the important role of the microbiota in this disease. Dietary composition and caloric intake appear to swiftly regulate intestinal microbial composition and function.SUMMARY: The interaction of the intestinal microbial world with its host, and its mutual regulation, will become one of the important topics of biomedical research and will provide us with further insights at the interface of microbiota, metabolism, metabolic syndrome, and obesity. A better understanding of the interaction between certain diets and the human gut microbiome should help to develop new guidelines for feeding humans at various time points in their life, help to improve global human health, and establish ways to prevent or treat various food-related diseases.KEYWORDS: gut microbiota, obesity, metabolic syndrome, type 2 diabetes
Styles APA, Harvard, Vancouver, ISO, etc.
4

Neophytou, Constantina, et Chrysoula Pitsouli. « How Gut Microbes Nurture Intestinal Stem Cells : A Drosophila Perspective ». Metabolites 12, no 2 (10 février 2022) : 169. http://dx.doi.org/10.3390/metabo12020169.

Texte intégral
Résumé :
Host-microbiota interactions are key modulators of host physiology and behavior. Accumulating evidence suggests that the complex interplay between microbiota, diet and the intestine controls host health. Great emphasis has been given on how gut microbes have evolved to harvest energy from the diet to control energy balance, host metabolism and fitness. In addition, many metabolites essential for intestinal homeostasis are mainly derived from gut microbiota and can alleviate nutritional imbalances. However, due to the high complexity of the system, the molecular mechanisms that control host-microbiota mutualism, as well as whether and how microbiota affects host intestinal stem cells (ISCs) remain elusive. Drosophila encompasses a low complexity intestinal microbiome and has recently emerged as a system that might uncover evolutionarily conserved mechanisms of microbiota-derived nutrient ISC regulation. Here, we review recent studies using the Drosophila model that directly link microbiota-derived metabolites and ISC function. This research field provides exciting perspectives for putative future treatments of ISC-related diseases based on monitoring and manipulating intestinal microbiota.
Styles APA, Harvard, Vancouver, ISO, etc.
5

Leardini, Davide, Edoardo Muratore, Daniele Zama, Arcangelo Prete, Andrea Pession et Riccardo Masetti. « Il ruolo del microbiota intestinale nella modulazione immunitaria ». Medico e Bambino pagine elettroniche 23, no 6 (30 juin 2020) : 130–36. http://dx.doi.org/10.53126/mebxxiiig130.

Texte intégral
Résumé :
The intestinal microbiota plays a crucial role in numerous physiological and pathological processes of the developmental age. The development of new investigation methods, such as next generation sequencing, has allowed a more precise characterization of the intestinal microbiota. This allowed us to deepen the complex relationship between the microbial flora and our organism. One of the many functions that emerged is the ability to modulate the host's immune system. The transplantation of hematopoietic stem cells represents a privileged setting for studying this interaction since, after the procedure, we witness the immune reconstruction starting from the donor precursors. The gut microbiota influences the development of major infectious and immune-mediated complications of transplantation and has a significant impact on patient survival. These evidences underline the possible therapeutic implications of the modulation of the intestinal microbiota. An in-depth study of the relationship between the immune system and microbiotic flora could allow us to better understand the role of the latter in other infectious or immune-mediated diseases.
Styles APA, Harvard, Vancouver, ISO, etc.
6

Пальцын, А. А. « Gut microbiota ». ZHurnal «Patologicheskaia fiziologiia i eksperimental`naia terapiia», no 4() (21 novembre 2018) : 202–8. http://dx.doi.org/10.25557/0031-2991.2018.04.202-208.

Texte intégral
Résumé :
Совершенствование методов генетического анализа и развернувшиеся на этой основе работы по международному проекту «Микробиом человека» представили большой объем знаний, существенно меняющих представления об эволюции живой природы и о механизмах развития многих болезней. Пришло осознание факта, что здоровье и болезни человека в большой степени определяются взаимодействием его собственных клеток с населяющими его тело (прежде всего кишечник) микробными (прежде всего бактериальными) клетками. Главенство в этом вопросе бактерий кишечника объясняется их подавляющей многочисленностью, позицией на первичном рубеже обмена со средой и невозможностью жизни без этого обмена. Результаты обширных теоретических и клинических исследований уже сегодня могут стать основой профилактики и терапии некоторых незаразных пандемий современности. Однако научно вполне достижимый дальнейший масштабный успех в преодолении этих пандемий тормозится внешними причинами. Кишечная микробиота связывает здоровье людей с экологией планеты, с сельским хозяйством и пищевой промышленностью. Действия же международных корпораций направляются интересами не здравоохранения, а получения максимальной выгоды в минимальные сроки. Improving methods of genetic analysis and the work developed on this basis under the «Human Microbiome» international project presented a large amount of knowledge that has significantly changed ideas about the evolution of living nature and mechanisms underlying development of many diseases. People became aware of the fact that human health and diseases are largely determined by the interaction of the body’s own cells with the microbial (primarily bacterial) cells inhabiting the body (primarily the intestine). The dominance of intestinal bacteria in this aspect is explained by their overwhelming multiplicity, position on the primary border of exchange with the environment, and the impossibility of life without this exchange. Results of extensive theoretical and clinical research already today can become a basis for prevention and treatment of some current non-infectious pandemics. However, although further, large-scale success in overcoming these pandemics is scientifically quite achievable, certain political reasons hamper it. Intestinal microbiota connects human health with the global ecology, agriculture, and the food industry while actions of governments and international corporations are driven by obtaining maximum benefits in the shortest possible time rather than interests of health care.
Styles APA, Harvard, Vancouver, ISO, etc.
7

Eom, Jung A., Goo Hyun Kwon, Na Yeon Kim, Eun Ju Park, Sung Min Won, Jin Ju Jeong, Ganesan Raja et al. « Diet-Regulating Microbiota and Host Immune System in Liver Disease ». International Journal of Molecular Sciences 22, no 12 (13 juin 2021) : 6326. http://dx.doi.org/10.3390/ijms22126326.

Texte intégral
Résumé :
The gut microbiota has been known to modulate the immune responses in chronic liver diseases. Recent evidence suggests that effects of dietary foods on health care and human diseases are related to both the immune reaction and the microbiome. The gut-microbiome and intestinal immune system play a central role in the control of bacterial translocation-induced liver disease. Dysbiosis, small intestinal bacterial overgrowth, translocation, endotoxemia, and the direct effects of metabolites are the main events in the gut-liver axis, and immune responses act on every pathways of chronic liver disease. Microbiome-derived metabolites or bacteria themselves regulate immune cell functions such as recognition or activation of receptors, the control of gene expression by epigenetic change, activation of immune cells, and the integration of cellular metabolism. Here, we reviewed recent reports about the immunologic role of gut microbiotas in liver disease, highlighting the role of diet in chronic liver disease.
Styles APA, Harvard, Vancouver, ISO, etc.
8

Janczy, Agata, Magdalena Landowska et Zdzisław Kochan. « Gut microbiome dysbiosis in anorexia nervosa ». Postępy Higieny i Medycyny Doświadczalnej 75 (29 avril 2021) : 283–91. http://dx.doi.org/10.5604/01.3001.0014.8601.

Texte intégral
Résumé :
Anorexia nervosa (AN) is described as an eating disorder, which is characterized by malnutrition, a fear of gaining body mass, and a disturbed self-body image. This disease is dependent on biological, psychological and socio-cultural factors. Among the various biological factors, the importance of intestinal microbiota has recently attracted much attention. Identification of the gut microbiota dysbiosis in patients with AN has opened new and promising research directions. Recent observations focus in particular on the association between intestinal microorganisms and the occurrence of functional gastrointestinal disorders associated with anorexia, anxiety and depression, as well as the regulation of eating habits. The composition of the gut microbiota differs between patients with AN and individuals with normal body mass. This is due to the incorrect diet of patients; on the other hand, there is growing interest in the role of intestinal microbiota in the pathogenesis of AN, its changes through re-nutrition practices, and in particular the modulation of intestinal microbiological composition by means of nutritional interventions or the use of preand probiotics as standard supplements therapy of eating disorders. There is a need for further research about the microbiome - intestine - brain axis. Furthermore, consequences of changes in dietary habits as part of AN treatment are also unknown. However, better knowledge about the relationship between the gut microbiome and the brain can help improve the treatment of this disorder. This review aims to present the current knowledge about the potential role of intestinal microbiota in the pathogenesis, course and treatment of AN.
Styles APA, Harvard, Vancouver, ISO, etc.
9

Chernevskaya, E. A., et N. V. Beloborodova. « Gut Microbiome in Critical Illness (Review) ». General Reanimatology 14, no 5 (28 octobre 2018) : 96–119. http://dx.doi.org/10.15360/1813-9779-2018-5-96-119.

Texte intégral
Résumé :
Radical changes in the composition, diversity and metabolic activity of gut microbiome in critically ill patients most probably affect adversely the outcome of treatment. Microbiota dysfunction may be a predictor and presumably the main cause of infectious complications and sepsis. Clinicists use objective scales for evaluation of patient condition severity including specific parameters of disorders of organs and systems; however, microbiota function is not considered specific and, hence, not evaluated. Technical capabilities of the recent decade have allowed characterizing the intestinal microbiota and that helped understanding the ongoing processes. The authors have analyzed data about the role of intestinal microbiota as a metabolic 'reactor' during critical states, possible complications related to misbalance of 'harmful' and 'beneficial' bacteria, and examined potential of a targeted therapy aimed directly at correction of intestinal microbiota. Search for papers was carried out using Scopus and Web of Science databases 2001 to 2018 years: (Gut Microbiota) AND (Critically ill OR Intensive care unit), key words taken for the search were: intestinal microbiota, metabolism, sepsis, antibiotics, critically ill patients, multiple organ failure. A number of questions in understanding of the interaction between gut microbiome and host remain open. It is necessary to take into account interference of microbial metabolism while assessing metabolome of patients with sepsis. Among low-molecular compounds found in blood of sepsis patients, special attention should be paid to molecules that can be classified as ‘common metabolites’ of humans and bacteria, for example, degradation products of aromatic compounds, which many-fold rise in blood of septic patients. It is necessary to take into consideration and experimentally model changes in the human internal environment, which occur during radical transformation of microbiome in critically ill patients. Such approach brings in new prospects for objective monitoring of diseases by evaluating metabolic profile at a particular moment of time based on integral indices reflecting the status of microbiome/metabolome system, which will supply new targets for therapeutic intervention in future.
Styles APA, Harvard, Vancouver, ISO, etc.
10

Garrett, Wendy S. « Gut Microbiota and Intestinal Inflammation ». Blood 120, no 21 (16 novembre 2012) : SCI—49—SCI—49. http://dx.doi.org/10.1182/blood.v120.21.sci-49.sci-49.

Texte intégral
Résumé :
Abstract Abstract SCI-49 Numerous studies of the microbiota that are found throughout the human body are under way, with the goal of unraveling the role of microbes in human physiology. Using experimental colitis models, we are investigating microbes that may instigate chronic inflammation, and we are studying putative beneficial microbes whose reduced presence may impact not only host response to the microbiota but also the behavior of the endogenous microbiota. Genomic approaches, combined with following microbial community response to a spectrum of dietary and pharmacologic perturbations, are shedding light on the dynamic operations of the microbiota that influence health and disease. Chronic inflammation in the intestine is not only the central pathophysiologic mechanism of inflammatory bowel disease (IBD) but also a key contributor to colorectal cancer. Ongoing work on the colorectal microbiome, using experimental models and human tumors, will be discussed. Collectively, our studies support the utility of wedding culture-independent and culture-dependent studies with mouse models for defining how the gut microbiota works in concert with the mucosal immune system to shape disease susceptibility for IBD and colorectal cancer. Disclosures: Garrett: Groupe Danone: Research Funding.
Styles APA, Harvard, Vancouver, ISO, etc.
11

Góralczyk-Bińkowska, Aleksandra, Dagmara Szmajda-Krygier et Elżbieta Kozłowska. « The Microbiota–Gut–Brain Axis in Psychiatric Disorders ». International Journal of Molecular Sciences 23, no 19 (24 septembre 2022) : 11245. http://dx.doi.org/10.3390/ijms231911245.

Texte intégral
Résumé :
Modulating the gut microbiome and its influence on human health is the subject of intense research. The gut microbiota could be associated not only with gastroenterological diseases but also with psychiatric disorders. The importance of factors such as stress, mode of delivery, the role of probiotics, circadian clock system, diet, and occupational and environmental exposure in the relationship between the gut microbiota and brain function through bidirectional communication, described as “the microbiome–gut–brain axis”, is especially underlined. In this review, we discuss the link between the intestinal microbiome and the brain and host response involving different pathways between the intestinal microbiota and the nervous system (e.g., neurotransmitters, endocrine system, immunological mechanisms, or bacterial metabolites). We review the microbiota alterations and their results in the development of psychiatric disorders, including major depressive disorder (MDD), schizophrenia (SCZ), bipolar disorder (BD), autism spectrum disorder (ASD), and attention-deficit hyperactivity disorder (ADHD).
Styles APA, Harvard, Vancouver, ISO, etc.
12

Vandenplas, Y. « Healthy gut microbiota and long term health ». Beneficial Microbes 6, no 2 (1 janvier 2015) : 173–79. http://dx.doi.org/10.3920/bm2014.0072.

Texte intégral
Résumé :
This review summarises how the composition of the gastro-intestinal microbiota depends on pre- and postnatal factors, and birth itself. The impact of method of delivery, feeding during infancy and medications, such as antibiotics and anti-acid medication, on the composition of the gastro-intestinal microbiota has clearly been shown. However, the duration of the impact of these factors is not well established. The gastro-intestinal microbiome composition is associated with many auto-immune mediated diseases. Although causality has not been obviously demonstrated, there is a strong tendency in this direction. Nevertheless, results of the manipulation of the gastro-intestinal microbiome composition in these conditions are often disappointing. A better understanding on factors determining the longterm composition of the gastro-intestinal microbiome and its health consequences are a priority research topic. A better understanding of the association between the microbiome and the immune system may have a tremendous impact on general health.
Styles APA, Harvard, Vancouver, ISO, etc.
13

Leblhuber, Friedrich, Daniela Ehrlich, Kostja Steiner, Simon Geisler, Dietmar Fuchs, Lukas Lanser et Katharina Kurz. « The Immunopathogenesis of Alzheimer’s Disease Is Related to the Composition of Gut Microbiota ». Nutrients 13, no 2 (25 janvier 2021) : 361. http://dx.doi.org/10.3390/nu13020361.

Texte intégral
Résumé :
The microbiota–gut–brain axis plays an important role in the development of neurodegenerative diseases. Commensal and pathogenic enteric bacteria can influence brain and immune system function by the production of lipopolysaccharides and amyloid. Dysbiosis of the intestinal microbiome induces local and consecutively systemic immune-mediated inflammation. Proinflammatory cytokines then trigger neuroinflammation and finally neurodegeneration. Immune-mediated oxidative stress can lead to a deficiency of vitamins and essential micronutrients. Furthermore, the wrong composition of gut microbiota might impair the intake and metabolization of nutrients. In patients with Alzheimer’s disease (AD) significant alterations of the gut microbiota have been demonstrated. Standard Western diet, infections, decreased physical activity and chronic stress impact the composition and diversity of gut microbiota. A higher abundancy of “pro-inflammatory” gut microbiota goes along with enhanced systemic inflammation and neuroinflammatory processes. Thus, AD beginning in the gut is closely related to the imbalance of gut microbiota. Modulation of gut microbiota by Mediterranean diet, probiotics and curcumin can slow down cognitive decline and alter the gut microbiome significantly. A multi-domain intervention approach addressing underlying causes of AD (inflammation, infections, metabolic alterations like insulin resistance and nutrient deficiency, stress) appears very promising to reduce or even reverse cognitive decline by exerting positive effects on the gut microbiota.
Styles APA, Harvard, Vancouver, ISO, etc.
14

Wang, Xinzhou, Peng Zhang et Xin Zhang. « Probiotics Regulate Gut Microbiota : An Effective Method to Improve Immunity ». Molecules 26, no 19 (8 octobre 2021) : 6076. http://dx.doi.org/10.3390/molecules26196076.

Texte intégral
Résumé :
Probiotics are beneficial active microorganisms that colonize the human intestines and change the composition of the flora in particular parts of the host. Recently, the use of probiotics to regulate intestinal flora to improve host immunity has received widespread attention. Recent evidence has shown that probiotics play significant roles in gut microbiota composition, which can inhibit the colonization of pathogenic bacteria in the intestine, help the host build a healthy intestinal mucosa protective layer, and enhance the host immune system. Based on the close relationship between the gut microbiota and human immunity, it has become an extremely effective way to improve human immunity by regulating the gut microbiome with probiotics. In this review, we discussed the influence of probiotics on the gut microbiota and human immunity, and the relationship between immunity, probiotics, gut microbiota, and life quality. We further emphasized the regulation of gut microflora through probiotics, thereby enhancing human immunity and improving people’s lives.
Styles APA, Harvard, Vancouver, ISO, etc.
15

Gupta, Haripriya, Gi Soo Youn, Min Jea Shin et Ki Tae Suk. « Role of Gut Microbiota in Hepatocarcinogenesis ». Microorganisms 7, no 5 (5 mai 2019) : 121. http://dx.doi.org/10.3390/microorganisms7050121.

Texte intégral
Résumé :
Hepatocellular carcinoma (HCC), one of the leading causes of death worldwide, has a causal nexus with liver injury, inflammation, and regeneration that accumulates over decades. Observations from recent studies have accounted for the involvement of the gut–liver axis in the pathophysiological mechanism responsible for HCC. The human intestine nurtures a diversified colony of microorganisms residing in the host ecosystem. The intestinal barrier is critical for conserving the normal physiology of the gut microbiome. Therefore, a rupture of this barrier or dysbiosis can cause the intestinal microbiome to serve as the main source of portal-vein endotoxins, such as lipopolysaccharide, in the progression of hepatic diseases. Indeed, increased bacterial translocation is a key sign of HCC. Considering the limited number of clinical studies on HCC with respect to the microbiome, we focus on clinical as well as animal studies involving the gut microbiota, with the current understandings of the mechanism by which the intestinal dysbiosis promotes hepatocarcinogenesis. Future research might offer mechanistic insights into the specific phyla targeting the leaky gut, as well as microbial dysbiosis, and their metabolites, which represent key pathways that drive HCC-promoting microbiome-mediated liver inflammation and fibrosis, thereby restoring the gut barrier function.
Styles APA, Harvard, Vancouver, ISO, etc.
16

Rinninella, Emanuele, Pauline Raoul, Marco Cintoni, Marta Palombaro, Gabriele Pulcini, Antonio Gasbarrini et Maria Cristina Mele. « Nutritional Interventions Targeting Gut Microbiota during Cancer Therapies ». Microorganisms 9, no 7 (9 juillet 2021) : 1469. http://dx.doi.org/10.3390/microorganisms9071469.

Texte intégral
Résumé :
The gut microbiome is increasingly being recognized for its influence on intestinal and extra-intestinal disorders such as cancer. Today, diet is the most studied environmental modulator of gut microbiota, capable of altering or improving it in terms of richness and diversity. Recent evidence from several preclinical and clinical trials suggested that gut microbiota composition could modulate cancer therapies (toxicities, treatment responses) and vice versa. This review highlights the latest research on the bidirectional associations between gut microbiota and cancer. We also dissect the role of gut microbiota during cancer therapies in terms of toxicity and treatment response and, in turn, how cancer therapies could impact gut microbiota composition and functions. In this context, we summarize the state-of-the-art research regarding the role of various nutritional interventions—prebiotics, dietary strategies, and dietary restrictions—as cutting-edge possibilities to modulate gut microbiota during cancer therapies.
Styles APA, Harvard, Vancouver, ISO, etc.
17

Kobayashi, Takashi, Michihiro Iwaki, Atsushi Nakajima, Asako Nogami et Masato Yoneda. « Current Research on the Pathogenesis of NAFLD/NASH and the Gut–Liver Axis : Gut Microbiota, Dysbiosis, and Leaky-Gut Syndrome ». International Journal of Molecular Sciences 23, no 19 (2 octobre 2022) : 11689. http://dx.doi.org/10.3390/ijms231911689.

Texte intégral
Résumé :
Global lifestyle changes have led to an increased incidence of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), requiring further in-depth research to understand the mechanisms and develop new therapeutic strategies. In particular, high-fat and high-fructose diets have been shown to increase intestinal permeability, which can expose the liver to endotoxins. Indeed, accumulating evidence points to a link between these liver diseases and the intestinal axis, including dysbiosis of the gut microbiome and leaky-gut syndrome. Here, we review the mechanisms contributing to these links between the liver and small intestine in the pathogenesis of NAFLD/NASH, focusing on the roles of intestinal microbiota and their metabolites to influence enzymes essential for proper liver metabolism and function. Advances in next-generation sequencing technology have facilitated analyses of the metagenome, providing new insights into the roles of the intestinal microbiota and their functions in physiological and pathological mechanisms. This review summarizes recent research linking the gut microbiome to liver diseases, offering new research directions to elucidate the detailed mechanisms and novel targets for treatment and prevention.
Styles APA, Harvard, Vancouver, ISO, etc.
18

Amir, Mohammed, katherine sanidad, Aparna Ananthanarayanan, Lingzhi Zhang, Nicholas Shiland, Naohiro Inohara, Gabriel Nunez et Melody Zeng. « Gut microbiota-reactive IgG regulates gut microbiota development and immunity against enteric pathogens in early life ». Journal of Immunology 208, no 1_Supplement (1 mai 2022) : 59.04. http://dx.doi.org/10.4049/jimmunol.208.supp.59.04.

Texte intégral
Résumé :
Abstract The gut microbiota elicits antigen-specific IgG at steady state that cross-reacts to pathogenic Gram-negative Enterobacteriaceae to confer protection against systemic infection. The role of these IgG antibodies in the development of gut microbiome and immunity against enteric pathogens in early life, however, remains largely undefined. In this study, we demonstrate that gut microbiota-induced maternal IgG is transferred to the neonatal gut through maternal milk via the neonatal Fc receptor and confers critical protection by inhibiting colonization and attachment of C. rodentium to the mucosa in the neonatal gut. Enhanced neonatal immunity against oral C. rodentium infection was observed following maternal immunization with a gut microbiota-derived IgG antigen, outer-membrane protein A (OMP-A), or induction of protective IgG antibodies in germ-free mice after reconstitution with IgG-inducing gut bacteria. Furthermore, we generated a mouse model with complete deficiency in all IgG isotypes. IgG-deficient neonatal mice exhibited increased susceptibility to C. rodentium infection, altered gut microbiome and increased numbers of intestinal IL-17-producing γδ T cells driven by the gut microbiome at steady state. The altered gut microbiome persisted into adulthood in IgG KO mice and contributed to increased disease severity in DSS-induced colitis. Taken together, our studies have defined critical roles for gut microbiome-induced IgG antibodies in shaping the gut microbiome and immunity against Gram-negative enteric pathogens from early life to adulthood.
Styles APA, Harvard, Vancouver, ISO, etc.
19

Ojezele, M. O. « Microbiome : pharmacokinetics, pharmacodynamics and drug/xenobiotic interactions ». African Journal of Clinical and Experimental Microbiology 21, no 2 (17 février 2020) : 78–87. http://dx.doi.org/10.4314/ajcem.v21i2.1.

Texte intégral
Résumé :
The participation of microbiota in myriads of physiological, metabolic, genetic and immunological processes shows that they are a fundamental part of human existence and health maintenance. The efficiency of drugs’ absorption depends on solubility, stability, permeability and metabolic enzymes produced by the body and gut microbiota. Two major types of microbiota-drug interaction have been identified; direct and indirect. The use of antibiotics is a direct means of targeting intestinal microbes and short-term use of antibiotic can significantly alter the microbiome composition. It is noteworthy that not every microbial drug metabolism is of benefit to the host as some drugs can shut down microbial processes as observed in the co-administration of antiviral sorivudine with fluoropyridimide resulting in a toxic buildup of fluoropyridimide metabolites from blockade of host fluoropyridimide by the microbial-sorivudine metabolite. It has been reported that many classes of drugs and xenobiotics modify the gut microbiome composition which may be detrimental to human health. Microbiome-drug interaction may be beneficial or detrimental resulting in either treatment success or failure which is largely dependent on factors such as microbial enzymes, chemical composition of candidate drug, host immunity and the complex relationship that exists with the microbiome. The effects of microbiota on pharmacology of drugs and vice versa are discussed in this review.Keywords: microbiome; pharmacokinetic, pharmacodynamic, drug, xenobiotic English Title: Microbiome: pharmacocinétique, pharmacodynamique et interactions médicamenteuses/xénobiotiquesLa participation du microbiote à des myriades de processus physiologiques, métaboliques, génétiques et immunologiques montre qu’ils sont un élément fondamental de l’existence et du maintien de la santé de l’être humain. L’efficacité de l’absorption des médicaments dépend de la solubilité, de la stabilité, de la perméabilité et des enzymes métaboliques produites par le corps et le microbiote intestinal. Deux types principaux d’interaction microbiote-médicament ont été identifiés; direct et indirect. L'utilisation d'antibiotiques est un moyen direct de cibler les microbes intestinaux et une utilisation à court terme d'antibiotique peut modifier de manière significative la composition du microbiome. Il est à noter que tous les métabolismes de médicaments microbiens ne sont pas bénéfiques pour l'hôte, car certains médicaments peuvent arrêter les processus microbiens observés lors de l'administration concomitante d'antiviral sorivudine et de fluoropyridimide, ce qui entraîne une accumulation toxique de métabolites de fluoropyridimide résultant du blocage du fluoropyridimide par l'hôte. métabolite microbien-sorivudine. Il a été rapporté que de nombreuses classes de médicaments et de xénobiotiques modifiaient la composition du microbiome intestinal, ce qui pourrait nuire à la santé humaine. Une interaction médicamenteuse-microbiome peut être bénéfique ou préjudiciable, entraînant le succès ou l'échec du traitement, qui dépend en grande partie de facteurs tels que les enzymes microbiennes, la composition chimique du médicament candidat, l'immunité de l'hôte et la relation complexe qui existe avec le microbiome. Les effets du microbiote sur la pharmacologie des médicaments et inversement sont discutés dans cette revue.Mots-clés: microbiome; pharmacocinétique, pharmacodynamique, médicament, xénobiotique
Styles APA, Harvard, Vancouver, ISO, etc.
20

Kalogeropoulos, Dimitrios, Konstantinos Katsikatsos, Konstantinos Dallas, Soon Wai Ch’ng, Ioannis Asproudis, Maria Stefaniotou et Chris Kalogeropoulos. « Role of intestinal microbiome in the pathogenesis of age-related macular degeneration ». Medical Hypothesis, Discovery & ; Innovation in Optometry 1, no 1 (31 août 2020) : 29–36. http://dx.doi.org/10.51329/mehdioptometry105.

Texte intégral
Résumé :
Background: The microbiome is strongly linked to many extra-intestinal disorders. Gut commensal microbiota, in particular, plays an active role in human immune and intestinal homeostasis. Complex interactions of the microbiota with host genetics and other underlying factors lead to intestinal dysbiosis, which is thought to be linked to ocular inflammatory diseases. Thus, the aim of this review is to analyze the role of intestinal microbiome in age-related macular degeneration (AMD). Methods: A thorough literature search was performed using PubMed/MEDLINE, limited to English language publications, from January 2004 to March 2020. An additional search was made employing Google Scholar to complete the collected data as per the above-mentioned time-line and language limitations. The main keywords used included age-related macular degeneration, microbiome, dysbiosis, autoimmunity, gut microbiota, epigenetics, immune-mediated inflammatory diseases, and gut-retina axis. Results: Recent studies have proposed the role of intestinal microbiota in the pathogenesis of AMD. Changes in the microbiome have been shown to trigger several ocular inflammatory processes. There is increasing evidence demonstrating that intestinal microbial imbalance may play an important role in the pathogenesis of AMD. Conclusions: This review summarizes how alterations in the intestinal microbiota can be associated with the pathogenesis of AMD and how new therapeutic modalities can be designed to target this microbiome to limit the severe nature of this disease. Future advances in microbiome research may unveil a new era in understanding and managing AMD.
Styles APA, Harvard, Vancouver, ISO, etc.
21

Zając, Marlena, Monika Borowiecka, Dariusz Gruca, Dagmara Buksak et Wiktor Wróblewski. « Gut Microbiome as a Novel Treatment Strategy for Psoriasis ». Journal of Education, Health and Sport 12, no 9 (3 septembre 2022) : 407–21. http://dx.doi.org/10.12775/jehs.2022.12.09.046.

Texte intégral
Résumé :
Introduction and purpose: Psoriasis is a skin disease that develops following chronic inflammatory signaling and keratinocyte hyperproliferation. The pathogenesis of psoriasis is compound and not yet fully understood. Several studies concerning gut microbiota composition and its role in disease pathogenesis recently demonstrated significant alterations among psoriatic patients. This study aims to highlight the latest scientific evidence regarding the gut microbiome alterations of psoriatic patients, as well as the state of knowledge in terms of microbiome-targeted therapies as promising preventive and therapeutic tools for psoriasis. Brief description of the state of knowledge: The current state of knowledge indicates that the main causes of psoriasis may be a genetic predisposition, as well as many immunological and environmental factors, including dysbiosis of the intestinal microflora. The article covers clinical and experimental studies which indicate that gut microbiota dysbiosis concerning diversity as well as the composition of the microbiome is the potential causal factor of psoriasis and the gut microbiota may serve as a promising prevention/therapy target for psoriasis patients. Conclusions: This review highlighted a strong link between psoriasis and the gut microbiota, to add new knowledge for discovering the relationship between the altered intestinal microbiota in psoriasis patients. Despite all of these interesting findings, there are a lot of limitations and challenges that future studies should face. More precise and greater studies need to be done to fully understand the potential of microbiota-aimed therapies.
Styles APA, Harvard, Vancouver, ISO, etc.
22

Kulchavenya, Ekaterina V. « Minerals contributing to human health and well-being ». Clinical review for general practice 2, no 1 (20 février 2021) : 58–64. http://dx.doi.org/10.47407/kr2021.2.1.00033.

Texte intégral
Résumé :
The gut microbiome is vital for normal human body functioning. The etiological and pathogenetic significance of increased intestinal permeability in disorders of various organs and systems seems to be certain. The term “microbiota-gut-brain axis” has been defined; the crucial role of the microbiota-gut-brain axis in neurological disorders has been confirmed. Gut microbiome not only contributes to digestion, metabolism and immunity, but also mediates sleep and mental health of the host via microbiota-gut-brain axis. Such elements as zinc and selenium are essential to maintain the microbial balance in the gut. Zinc contributes to gut homeostasis and Paneth cell function. Zinc has a direct impact on gut microbiota composition (for example, on some species of Staphylococcus), modulates gut microbiota reducing the fatal entry of bacteria into the bloodstream and lymphatic vessels. Thus, zinc alters microbiome due to direct cytotoxic / cytostatic effect on certain bacteria, such as staphylococci. Zinc possesses therapeutic effect in gastrointestinal infections and diarrhea. Bacterial translocation may be also reduced with Rebamipide possessing cytoprotective and antioxidant activity. Selenium in the form of selenoproteins has a number of functions in normal health and metabolism. Selenium contributes to immune system functioning and to progression of HIV to AIDS. Selenium deficiency results in cardiovascular diseases, infertility, myodegenerative disorders, and cognitive decline.
Styles APA, Harvard, Vancouver, ISO, etc.
23

Gallo, Benjamin D., John M. Farrell et Brian Leydet. « Use of next generation sequencing to compare simple habitat and species level differences in the gut microbiota of an invasive and native freshwater fish species ». PeerJ 8 (18 décembre 2020) : e10237. http://dx.doi.org/10.7717/peerj.10237.

Texte intégral
Résumé :
Research on the gut microbiome of host organisms has rapidly advanced with next generation sequencing (NGS) and high-performance computing capabilities. Nonetheless, gut microbiome research has focused on mammalian organisms in laboratory settings, and investigations pertaining to wild fish gut microbiota remain in their infancy. We applied a procedure (available at https://github.com/bngallo1994) for sampling of the fish gut for use in NGS to describe microbial community structure. Our approach allowed for high bacterial OTU diversity coverage (>99.7%, Good’s Coverage) that led to detection of differences in gut microbiota of an invasive (Round Goby) and native (Yellow Bullhead) fish species and collected from the upper St. Lawrence River, an environment where the gut microbiota of fish had not previously been tested. Additionally, results revealed habitat level differences in gut microbiota using two distance metrics (Unifrac, Bray–Curtis) between nearshore littoral and offshore profundal collections of Round Goby. Species and habitat level differences in intestinal microbiota may be of importance in understanding individual and species variation and its importance in regulating fish health and physiology.
Styles APA, Harvard, Vancouver, ISO, etc.
24

Eindor-Abarbanel, Adi, Genelle R. Healey et Kevan Jacobson. « Therapeutic Advances in Gut Microbiome Modulation in Patients with Inflammatory Bowel Disease from Pediatrics to Adulthood ». International Journal of Molecular Sciences 22, no 22 (19 novembre 2021) : 12506. http://dx.doi.org/10.3390/ijms222212506.

Texte intégral
Résumé :
There is mounting evidence that the gut microbiota plays an important role in the pathogenesis of inflammatory bowel disease (IBD). For the past decade, high throughput sequencing-based gut microbiome research has identified characteristic shifts in the composition of the intestinal microbiota in patients with IBD, suggesting that IBD results from alterations in the interactions between intestinal microbes and the host’s mucosal immune system. These studies have been the impetus for the development of new therapeutic approaches targeting the gut microbiome, such as nutritional therapies, probiotics, fecal microbiota transplant and beneficial metabolic derivatives. Innovative technologies can further our understanding of the role the microbiome plays as well as help to evaluate how the different approaches in microbiome modulation impact clinical responses in adult and pediatric patients. In this review, we highlight important microbiome studies in patients with IBD and their response to different microbiome modulation therapies, and describe the differences in therapeutic response between pediatric and adult patient cohorts.
Styles APA, Harvard, Vancouver, ISO, etc.
25

Chen, Grace. « The Role of the Gut Microbiome in Colorectal Cancer ». Clinics in Colon and Rectal Surgery 31, no 03 (1 avril 2018) : 192–98. http://dx.doi.org/10.1055/s-0037-1602239.

Texte intégral
Résumé :
AbstractThere is increasing evidence that the gut microbiome, which consists of trillions of microbes representing over 1,000 species of bacteria with over 3 million genes, significantly impacts intestinal health and disease. The gut microbiota not only is capable of promoting intestinal homeostasis and antitumor responses but can also contribute to chronic dysregulated inflammation as well as have genotoxic effects that lead to carcinogenesis. Whether the gut microbiota maintains health or promotes colon cancer may ultimately depend on the composition of the gut microbiome and the balance within the microbial community of protective and detrimental bacterial populations. Disturbances in the normal balanced state of a healthful microbiome, known as dysbiosis, have been observed in patients with colorectal cancer (CRC); however, whether these alterations precede and cause CRC remains to be determined. Nonetheless, studies in mice strongly suggest that the gut microbiota can modulate susceptibility to CRC, and therefore may serve as both biomarkers and therapeutic targets.
Styles APA, Harvard, Vancouver, ISO, etc.
26

Huang, Qin, Fang Yu, Di Liao et Jian Xia. « Microbiota-Immune System Interactions in Human Neurological Disorders ». CNS & ; Neurological Disorders - Drug Targets 19, no 7 (26 novembre 2020) : 509–26. http://dx.doi.org/10.2174/1871527319666200726222138.

Texte intégral
Résumé :
: Recent studies implicate microbiota-brain communication as an essential factor for physiology and pathophysiology in brain function and neurodevelopment. One of the pivotal mechanisms about gut to brain communication is through the regulation and interaction of gut microbiota on the host immune system. In this review, we will discuss the role of microbiota-immune systeminteractions in human neurological disorders. The characteristic features in the development of neurological diseases include gut dysbiosis, the disturbed intestinal/Blood-Brain Barrier (BBB) permeability, the activated inflammatory response, and the changed microbial metabolites. Neurological disorders contribute to gut dysbiosis and some relevant metabolites in a top-down way. In turn, the activated immune system induced by the change of gut microbiota may deteriorate the development of neurological diseases through the disturbed gut/BBB barrier in a down-top way. Understanding the characterization and identification of microbiome-immune- brain signaling pathways will help us to yield novel therapeutic strategies by targeting the gut microbiome in neurological disease.
Styles APA, Harvard, Vancouver, ISO, etc.
27

Singh, Samradhi, Poonam Sharma, Namrata Pal, Manoj Kumawat, Swasti Shubham, Devojit Kumar Sarma, Rajnarayan R. Tiwari, Manoj Kumar et Ravinder Nagpal. « Impact of Environmental Pollutants on Gut Microbiome and Mental Health via the Gut–Brain Axis ». Microorganisms 10, no 7 (19 juillet 2022) : 1457. http://dx.doi.org/10.3390/microorganisms10071457.

Texte intégral
Résumé :
Over the last few years, the microbiome has emerged as a high-priority research area to discover missing links between brain health and gut dysbiosis. Emerging evidence suggests that the commensal gut microbiome is an important regulator of the gut–brain axis and plays a critical role in brain physiology. Engaging microbiome-generated metabolites such as short-chain fatty acids, the immune system, the enteric nervous system, the endocrine system (including the HPA axis), tryptophan metabolism or the vagus nerve plays a crucial role in communication between the gut microbes and the brain. Humans are exposed to a wide range of pollutants in everyday life that impact our intestinal microbiota and manipulate the bidirectional communication between the gut and the brain, resulting in predisposition to psychiatric or neurological disorders. However, the interaction between xenobiotics, microbiota and neurotoxicity has yet to be completely investigated. Although research into the precise processes of the microbiota–gut–brain axis is growing rapidly, comprehending the implications of environmental contaminants remains challenging. In these milieus, we herein discuss how various environmental pollutants such as phthalates, heavy metals, Bisphenol A and particulate matter may alter the intricate microbiota–gut–brain axis thereby impacting our neurological and overall mental health.
Styles APA, Harvard, Vancouver, ISO, etc.
28

Kharchenko, Yu V., H. I. Titov, D. H. Kryzhanovskyi, M. P. Fedchenko, H. P. Chernenko, V. V. Filipenko et V. A. Miakushko. « Stress and the Gut-Brain Axis ». Ukraïnsʹkij žurnal medicini, bìologìï ta sportu 7, no 4 (30 août 2022) : 137–46. http://dx.doi.org/10.26693/jmbs07.04.137.

Texte intégral
Résumé :
The purpose of the review was to study the effects of stress on the gut microbiota. Results and discussion. The gut microbiota forms a complex microbial community that has a significant impact on human health. The composition of the microbiota varies from person to person, and it changes throughout life. It is known that the microbiome can be altered due to diet, various processes, such as inflammation and/or stress. Like all other areas of medicine, microbiology is constantly growing. The gut microbiota lives in a symbiotic relationship with the human host. It is now believed to interact with almost all human organs, including the central nervous system, in the so-called «gut-brain-microbiome axis». Recently, a growing level of research is showing that microbes play a much bigger role in our lives than previously thought, and can have a myriad of effects on how we behave and think, and even on our mental health. The relationship between the brain and the microbiota is bidirectional and includes endocrine, neuronal, immune, and metabolic pathways. The microbiota interacts with the brain through various mechanisms and mediators, including cytokines, short-chain fatty acids, hormones, and neurotransmitters. According to the hypothalamic-pituitary-adrenocortical axis imbalance theory, hormonal imbalances are closely related to psychiatric illness, anxiety, and stress disorders. Therefore, the gut microbiome is closely related to the development and functioning of this axis. The microbiota can influence neurotransmitter levels in a variety of ways, including the secretion of gamma-aminobutyric acid, norepinephrine, dopamine, and serotonin, and can even regulate serotonin synthesis. These neurotransmitters can influence the hormonal status of the body, and the hormones themselves can influence the formation of the qualitative and quantitative composition of the microbiota. Accordingly, a change in the composition of the intestinal microbiota may be responsible for modifying the hormonal levels of the human body. The endocrine environment in the gut can also be modulated through the neuro-enteroendocrine system. Conclusion. Today, it is known that microbiota changes can be associated with several disorders of the nervous system, such as neuropsychiatric, neurodegenerative and neuroinflammatory processes. Research in recent decades has shown that disorders of the nervous system and mood disorders are associated with changes in the balance of neurotransmitters in the brain. Therefore, understanding the role of microbiota in the development and functioning of the brain is of great importance
Styles APA, Harvard, Vancouver, ISO, etc.
29

Bao, Xiaowei, Dongwen Bai, Xiaolu Liu, Ying Wang, Lanjun Zeng, Chenye Wei et Weiquan Jin. « Effects of the Cistanche tubulosa Aqueous Extract on the Gut Microbiota of Mice with Intestinal Disorders ». Evidence-Based Complementary and Alternative Medicine 2021 (14 juillet 2021) : 1–11. http://dx.doi.org/10.1155/2021/4936970.

Texte intégral
Résumé :
Disorders of the gut microbiota are associated with many diseases. The aqueous extract from Cistanche tubulosa (CT), a traditional Chinese herbal formula, has been reported to play a role in protecting the human intestine. However, little is known about its effects on the gut microbiota. The present study was carried out to determine whether the CT aqueous extract can modulate the gut microbiome in mice with intestinal disorders. We found that the damaged intestinal morphology resulting from treatment with cefixime could be rescued using the CT aqueous extract. The comparison of microbial diversity between mice treated with the CT extract and control mice also indicated that the disorder in the microbiome community of model groups could be restored by treatment with high and medium concentrations of the CT aqueous extract. Treatment with cefixime led to a significant decrease in lactic acid bacteria; however, the supplementation of the CT aqueous extract recovered the growth of these lactic acid bacteria. Furthermore, the CT aqueous extract was able to moderate the dramatic changes in the metabolic pathways of the gut microbiome induced by cefixime. These findings provided an insight into the beneficial effects of the CT aqueous extract on gut microbiota, and they also provided an important reference for the development of related drugs in the future.
Styles APA, Harvard, Vancouver, ISO, etc.
30

Yang, Lina, Li Li, Xinghui Wu, Wenqi Cai, Qian Lin, Danshi Zhu et He Liu. « The Effect of Natural Soluble Polysaccharides on the Type 2 Diabetes through Modulating Gut Microbiota : A Review ». Current Medicinal Chemistry 28, no 26 (8 septembre 2021) : 5368–85. http://dx.doi.org/10.2174/0929867328666210309110352.

Texte intégral
Résumé :
Diabetes strongly influences the patient’s quality of life. The incidence of type 2 diabetes (T2D) accounts for approximately 90% of diabetic patients. Natural polysaccharides have been widely used for diabetes management. Changes in gut microbiota can also be used for the prevention and treatment of diabetes. In this review, the effects of different natural polysaccharides on gut microbiota, as well as the relationship between diabetes and the gut microbiome are summarized. The intestine is the primary location in which natural polysaccharides exert their biological activities and plays an important role in maintaining healthy bodily functions. Polysaccharides change the composition of the gut microbiota, which inhibits pathogen invasion and promotes beneficial bacterial growth. In addition, the gut microbiota degrade polysaccharides and produce metabolites to further modify the intestinal environment. Interestingly, the metabolites (short chain fatty acids and other bioactive components) have been shown to improve gut health, control glycemia, lower lipids, reduce insulin resistance, and alleviate inflammation. Therefore, understanding the underlying mechanisms by which soluble polysaccharides improve T2D through regulating the gut microbiota and provide a future reference for the management of T2D and its associated complications.
Styles APA, Harvard, Vancouver, ISO, etc.
31

Ticinesi, Andrea, Antonio Nouvenne, Vincenzo Corrente, Claudio Tana, Francesco Di Mario et Tiziana Meschi. « Diverticular Disease : a Gut Microbiota Perspective ». Journal of Gastrointestinal and Liver Diseases 28, no 3 (1 septembre 2019) : 327–37. http://dx.doi.org/10.15403/jgld-277.

Texte intégral
Résumé :
Gut microbiota composition and functionality are involved in the pathophysiology of several intestinal andextraintestinal diseases, and are increasingly considered a modulator of local and systemic inflammation.However, the involvement of gut microbiota in diverticulosis and in diverticular disease is still poorlyinvestigated. In this review, we critically analyze the existing evidence on the fecal and mucosa-associatedmicrobiota composition and functionality across different stages of diverticular disease. We also explorethe influence of risk factors for diverticulosis on gut microbiota composition, and speculate on the possiblerelevance of these associations for the pathogenesis of diverticula. We overview the current treatments ofdiverticular disease targeting the intestinal microbiome, highlighting the current areas of uncertainty andthe need for future studies. Although no conclusive remarks on the relationship between microbiota anddiverticular disease can be made, preliminary data suggest that abdominal symptoms are associated withreduced representation of taxa with a possible anti-inflammatory effect, such as Clostridium cluster IV, andovergrowth of Enterobacteriaceae, Bifidobacteria and Akkermansia. The role of the microbiota in the earlystages of the disease is still very uncertain. Future studies should help to disentangle the role of the microbiomein the pathogenesis of diverticular disease and its progression towards more severe forms.
Styles APA, Harvard, Vancouver, ISO, etc.
32

Sadeghpour Heravi, Fatemah, et Honghua Hu. « Bifidobacterium : Host–Microbiome Interaction and Mechanism of Action in Preventing Common Gut-Microbiota-Associated Complications in Preterm Infants : A Narrative Review ». Nutrients 15, no 3 (30 janvier 2023) : 709. http://dx.doi.org/10.3390/nu15030709.

Texte intégral
Résumé :
The development and health of infants are intertwined with the protective and regulatory functions of different microorganisms in the gut known as the gut microbiota. Preterm infants born with an imbalanced gut microbiota are at substantial risk of several diseases including inflammatory intestinal diseases, necrotizing enterocolitis, late-onset sepsis, neurodevelopmental disorders, and allergies which can potentially persist throughout adulthood. In this review, we have evaluated the role of Bifidobacterium as commonly used probiotics in the development of gut microbiota and prevention of common diseases in preterm infants which is not fully understood yet. The application of Bifidobacterium as a therapeutical approach in the re-programming of the gut microbiota in preterm infants, the mechanisms of host-microbiome interaction, and the mechanism of action of this bacterium have also been investigated, aiming to provide new insights and opportunities in microbiome-targeted interventions in personalized medicine.
Styles APA, Harvard, Vancouver, ISO, etc.
33

Shmagel, Anna, Ryan Demmer, Daniel Knights, Mary Butler, Lisa Langsetmo, Nancy Lane et Kristine Ensrud. « The Effects of Glucosamine and Chondroitin Sulfate on Gut Microbial Composition : A Systematic Review of Evidence from Animal and Human Studies ». Nutrients 11, no 2 (30 janvier 2019) : 294. http://dx.doi.org/10.3390/nu11020294.

Texte intégral
Résumé :
Oral glucosamine sulfate (GS) and chondroitin sulfate (CS), while widely marketed as joint-protective supplements, have limited intestinal absorption and are predominantly utilized by gut microbiota. Hence the effects of these supplements on the gut microbiome are of great interest, and may clarify their mode of action, or explain heterogeneity in therapeutic responses. We conducted a systematic review of animal and human studies reporting the effects of GS or CS on gut microbial composition. We searched MEDLINE, EMBASE, and Scopus databases for journal articles in English from database inception until July 2018, using search terms microbiome, microflora, intestinal microbiota/flora, gut microbiota/flora and glucosamine or chondroitin. Eight original articles reported the effects of GS or CS on microbiome composition in adult humans (four articles) or animals (four articles). Studies varied significantly in design, supplementation protocols, and microbiome assessment methods. There was moderate-quality evidence for an association between CS exposure and increased abundance of genus Bacteroides in the murine and human gut, and low-quality evidence for an association between CS exposure and an increase in Desulfovibrio piger species, an increase in Bacteroidales S24-7 family, and a decrease in Lactobacillus. We discuss the possible metabolic implications of these changes for the host. For GS, evidence of effects on gut microbiome was limited to one low-quality study. This review highlights the importance of considering the potential influence of oral CS supplements on gut microbiota when evaluating their effects and safety for the host.
Styles APA, Harvard, Vancouver, ISO, etc.
34

Kohl, Hannah M., Andrea R. Castillo et Javier Ochoa-Repáraz. « The Microbiome as a Therapeutic Target for Multiple Sclerosis : Can Genetically Engineered Probiotics Treat the Disease ? » Diseases 8, no 3 (30 août 2020) : 33. http://dx.doi.org/10.3390/diseases8030033.

Texte intégral
Résumé :
There is an increasing interest in the intestinal microbiota as a critical regulator of the development and function of the immune, nervous, and endocrine systems. Experimental work in animal models has provided the foundation for clinical studies to investigate associations between microbiota composition and function and human disease, including multiple sclerosis (MS). Initial work done using an animal model of brain inflammation, experimental autoimmune encephalomyelitis (EAE), suggests the existence of a microbiota–gut–brain axis connection in the context of MS, and microbiome sequence analyses reveal increases and decreases of microbial taxa in MS intestines. In this review, we discuss the impact of the intestinal microbiota on the immune system and the role of the microbiome–gut–brain axis in the neuroinflammatory disease MS. We also discuss experimental evidence supporting the hypothesis that modulating the intestinal microbiota through genetically modified probiotics may provide immunomodulatory and protective effects as a novel therapeutic approach to treat this devastating disease.
Styles APA, Harvard, Vancouver, ISO, etc.
35

Buyvalenko, U. V., et E. V. Pokrovskaya. « Interaction between the gut microbiota and oral antihyperglycemic drugs ». Problems of Endocrinology 68, no 2 (20 janvier 2022) : 66–71. http://dx.doi.org/10.14341/probl12835.

Texte intégral
Résumé :
The gut microbiome is the largest microbial habitat in the human body. The main functions include obtaining energy from complex food fibers, maturation and formation of the immune system, intestinal angiogenesis, restoration of epithelial damage to the intestine, development of the nervous system, protection against pathogens, etc. It is also known that a number of drugs can cause changes in the composition of the intestinal microflora, and intestinal bacteria, in turn, produce a number of enzymes and metabolites that can chemically change the structure of drugs, leading to more side effects, and in some cases to positive changes. In this review we present current evidence supporting the effects of microbiota in host-drug interactions, in particular, the reciprocal effects of gut microbiota and oral hypoglycemic drugs on each other. Gaining and evaluating knowledge in this area will help pave the way for the development of new microbiota-based strategies that can be used in the future to improve treatment outcomes for type 2 diabetes mellitus (T2D).
Styles APA, Harvard, Vancouver, ISO, etc.
36

Wright, Michelle L., Christina Fournier, Madelyn C. Houser, Malú Tansey, Jonathan Glass et Vicki Stover Hertzberg. « Potential Role of the Gut Microbiome in ALS : A Systematic Review ». Biological Research For Nursing 20, no 5 (20 juin 2018) : 513–21. http://dx.doi.org/10.1177/1099800418784202.

Texte intégral
Résumé :
Amyotrophic lateral sclerosis (ALS) etiology and pathophysiology are not well understood. Recent data suggest that dysbiosis of gut microbiota may contribute to ALS etiology and progression. This review aims to explore evidence of associations between gut microbiota and ALS etiology and pathophysiology. Databases were searched for publications relevant to the gut microbiome in ALS. Three publications provided primary evidence of changes in microbiome profiles in ALS. An ALS mouse model revealed damaged tight junction structure and increased permeability in the intestine versus controls along with a shifted microbiome profile, including decreased levels of butyrate-producing bacteria. In a subsequent publication, again using an ALS mouse model, researchers showed that dietary supplementation with butyrate relieved symptoms and lengthened both time to onset of weight loss and survival time. In a small study of ALS patients and healthy controls, investigators also found decreased levels of butyrate-producing bacteria. Essential for maintaining gut barrier integrity, butyrate is the preferred energy source of intestinal epithelial cells. Ten other articles were reviews and commentaries providing indirect support for a role of gut microbiota in ALS pathophysiology. Thus, these studies provide a modicum of evidence implicating gut microbiota in ALS disease, although more research is needed to confirm the connection and determine pathophysiologic mechanisms. Nurses caring for these patients need to understand the gut microbiome and its potential role in ALS in order to effectively counsel patients and their families about emerging therapies (e.g., prebiotics, probiotics, and fecal microbial transplant) and their off-label uses.
Styles APA, Harvard, Vancouver, ISO, etc.
37

Fraga, A., D. Esteves-Sousa, J. Facucho-Oliveira, M. Albuquerque, M. Costa, P. Espada-Santos, N. Moura et A. Moutinho. « Mechanisms linking gut microbiota to depression ». European Psychiatry 64, S1 (avril 2021) : S741. http://dx.doi.org/10.1192/j.eurpsy.2021.1962.

Texte intégral
Résumé :
IntroductionThe gut microbiota constitute the largest and most diverse community in the body which is primarily responsible for the maintenance of the intestinal wall integrity and the protection against pathogens. Besides having an important role in the regulation of host energy metabolism, the gut microbiota can also influence neurodevelopment, modulate behavioral and might contribute to the development of psychiatry disorders.ObjectivesThe authors elaborated a narrative literature review to understand how gut microbiota can influence depression.MethodsUsing PubMed as the database, a research was conducted about how Gut Microbiota relates with Depression.ResultsThe microbiota-gut-brain axis encompasses the strong bidirectional communication between the gut microbiota and the CNS. Multiple mechanisms may be involved in this bilateral communication, including immune, endocrine and neural pathways. Permutations in the gut microbiome composition trigger microbial lipopolysaccharides production that activates inflammatory responses. Cytokines send signals to the vagus nerve, which links the process to the hypothalamic-pituitary-adrenal axis that consequently causes behavioral effects. Beyond this, gut microbiota have the capacity to produce many neurotransmitters and neuromodulators such as serotonin and can induce the secretion of the brain-derived neurotrophic factor, an important plasticity-related protein that promotes neuronal growth, development and survival.ConclusionsNeuroinflammatory processes like those that occur in depression are deeply modulated by peripheral inflammatory stimuli, especially those from the intestinal microbiota. However, the knowledge is currently limited and the information available is not enough to understand the exact mechanisms. Therefore, more studies are required to show how gut microbiota influences the human brain.DisclosureNo significant relationships.
Styles APA, Harvard, Vancouver, ISO, etc.
38

Gianchecchi, Elena, et Alessandra Fierabracci. « Recent Advances on Microbiota Involvement in the Pathogenesis of Autoimmunity ». International Journal of Molecular Sciences 20, no 2 (11 janvier 2019) : 283. http://dx.doi.org/10.3390/ijms20020283.

Texte intégral
Résumé :
Autoimmune disorders derive from genetic, stochastic, and environmental factors that all together interact in genetically predisposed individuals. The impact of an imbalanced gut microbiome in the pathogenesis of autoimmunity has been suggested by an increasing amount of experimental evidence, both in animal models and humans. Several physiological mechanisms, including the establishment of immune homeostasis, are influenced by commensal microbiota in the gut. An altered microbiota composition produces effects in the gut immune system, including defective tolerance to food antigens, intestinal inflammation, and enhanced gut permeability. In particular, early findings reported differences in the intestinal microbiome of subjects affected by several autoimmune conditions, including prediabetes or overt disease compared to healthy individuals. The present review focuses on microbiota-host homeostasis, its alterations, factors that influence its composition, and putative involvement in the development of autoimmune disorders. In the light of the existing literature, future studies are necessary to clarify the role played by microbiota modifications in the processes that cause enhanced gut permeability and molecular mechanisms responsible for autoimmunity onset.
Styles APA, Harvard, Vancouver, ISO, etc.
39

Golovach, I. Yu, et D. G. Rekalov. « Osteoarthritis and intestinal microbiota : pathogenetic significance of the joint — gut — microbiome axis ». PAIN, JOINTS, SPINE 12, no 2 (27 novembre 2022) : 72–80. http://dx.doi.org/10.22141/pjs.12.2.2022.332.

Texte intégral
Résumé :
Introduction. Osteoarthritis (ОА) is a disease leading to joint degeneration, accompanied by constant pain, inflammation, and functional failure of the joints. Although many factors contribute to the development of ОА, the gut microbiome has recently emerged as an important pathogenic factor in ОА initiation and progression. The purpose of the study was to analyze modern literature data regarding the link between the gut microbiome and ОА. Materials and me­thods. The available data of clinical studies and scientific reviews were analyzed, and modern meta-analyses on the influence of gut microbiota on the development and progression of ОА were evaluated. Results. Gut microbiota is responsible for a number of metabolic, immunolo­gical, and structural and neurological functions, potentially elucida­ting the heterogeneity of OA phenotypes and formation of individual features of the course of the disease. Numerous studies support the hypothesis of the existence of a gut – joint axis and the interaction between gut microbiota and OA-relevant risk factors. The proposed concept begins with intestinal disruption and dysbacteriosis, disruption of microbiota homeostasis, continuous changes in microbial composition and genomic plasticity for optimal adaptation of bacteria to the host environment, accompanied by both adaptive and innate immune responses due to translocation of bacteria and bacterial products into the bloodstream to the joint. This cascade ultimately leads to inflammation in the joint and contributes to the development and progression of OA. Interpretion of the potential mechanisms of OA pathogenesis is essential for the development of new preventive and disease-modifying therapeutic interventions. In addition, gut microbiota is also a potential biomarker related to inflammation and gut dysbiosis to predict the progression of ОА and monitor the effectiveness of therapeutic interventions. Conclusions. In this review, we summarized research data that are supporting the hypothesis of a “joint – gut – microbiota axis” and the interaction between gut microbiota and the OA-relevant factors, including age, gender, metabolism, obesity.
Styles APA, Harvard, Vancouver, ISO, etc.
40

Bhargava, Shruti, Erik Merckelbach, Heidi Noels, Ashima Vohra et Joachim Jankowski. « Homeostasis in the Gut Microbiota in Chronic Kidney Disease ». Toxins 14, no 10 (20 septembre 2022) : 648. http://dx.doi.org/10.3390/toxins14100648.

Texte intégral
Résumé :
The gut microbiota consists of trillions of microorganisms, fulfilling important roles in metabolism, nutritional intake, physiology and maturation of the immune system, but also aiding and abetting the progression of chronic kidney disease (CKD). The human gut microbiome consists of bacterial species from five major bacterial phyla, namely Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia. Alterations in the members of these phyla alter the total gut microbiota, with a decline in the number of symbiotic flora and an increase in the pathogenic bacteria, causing or aggravating CKD. In addition, CKD-associated alteration of this intestinal microbiome results in metabolic changes and the accumulation of amines, indoles and phenols, among other uremic metabolites, which have a feedforward adverse effect on CKD patients, inhibiting renal functions and increasing comorbidities such as atherosclerosis and cardiovascular diseases (CVD). A classification of uremic toxins according to the degree of known toxicity based on the experimental evidence of their toxicity (number of systems affected) and overall experimental and clinical evidence was selected to identify the representative uremic toxins from small water-soluble compounds, protein-bound compounds and middle molecules and their relation to the gut microbiota was summarized. Gut-derived uremic metabolites accumulating in CKD patients further exhibit cell-damaging properties, damage the intestinal epithelial cell wall, increase gut permeability and lead to the translocation of bacteria and endotoxins from the gut into the circulatory system. Elevated levels of endotoxins lead to endotoxemia and inflammation, further accelerating CKD progression. In recent years, the role of the gut microbiome in CKD pathophysiology has emerged as an important aspect of corrective treatment; however, the mechanisms by which the gut microbiota contributes to CKD progression are still not completely understood. Therefore, this review summarizes the current state of research regarding CKD and the gut microbiota, alterations in the microbiome, uremic toxin production, and gut epithelial barrier degradation.
Styles APA, Harvard, Vancouver, ISO, etc.
41

Hirano, Takehiro, et Hiroshi Nakase. « The Multifaceted Effects of Gut Microbiota on the Immune System of the Intestinal Mucosa ». Immuno 1, no 4 (13 décembre 2021) : 583–94. http://dx.doi.org/10.3390/immuno1040041.

Texte intégral
Résumé :
The gut microbiota has diverse microbial components, including bacteria, viruses, and fungi. The interaction between gut microbiome components and immune responses has been studied extensively over the last decade. Several studies have reported the potential role of the gut microbiome in maintaining gut homeostasis and the development of disease. The commensal microbiome can preserve the integrity of the mucosal barrier by acting on the host immune system. Contrastingly, dysbiosis-induced inflammation can lead to the initiation and progression of several diseases through inflammatory processes and oxidative stress. In this review, we describe the multifaceted effects of the gut microbiota on several diseases from the perspective of mucosal immunological responses.
Styles APA, Harvard, Vancouver, ISO, etc.
42

Fetarayani, Deasy, Handoko Hariyono et Gatot Soegiarto. « The role of gut microbiota in health and diseases ». Qanun Medika - Medical Journal Faculty of Medicine Muhammadiyah Surabaya 5, no 1 (26 janvier 2021) : 19. http://dx.doi.org/10.30651/jqm.v5i1.5846.

Texte intégral
Résumé :
ABSTRACTABSTRACTMicrobiota contributes a crucial part in the human hosts' health and actively provides to the emergence of various diseases. The optimal composition of healthy intestinal microbiota varies from person to person. The more various and abundant of the microbiota, the greater their resistance to outside hazards. Colonization of the microbiota in the human body starts after delivery and develops continuously from infant to adult. The largest microbial colony is constructed in the lower part of the adult human digestive tract. The composition of the human intestinal microbiota alters promptly during the beginning of life and is steady. It has been described the close relationship among dysbiosis of the intestinal microbiota with intestinal and non-intestinal diseases. Nevertheless, it is uncertain whether dysbiosis is the culprit of the disease or only as a result of the disease. Human microbiota's role must be investigated more deeply so that later it can be developed for the prevention, diagnosis of disease, and more effective treatment strategies in the future. In this minireview, we will describe the development of the gut microbiota, its interaction with our bodily systems and defense, the multiple causes of dysbiosis, and its impact on several metabolic in inflammatory diseases in humans. With this insight, it is hoped that we can be more cautious about using antibiotics, avoid things that lead to dysbiosis, and handle diseases more holistically, putting the balance of the microbiota into account. Keywords: human, microbiota, gastrointestinal tract, dysbiosis, health and diseaseCorresponding author: deasyfetarayani@gmail.com
Styles APA, Harvard, Vancouver, ISO, etc.
43

Doroszkiewicz, Julia, Magdalena Groblewska et Barbara Mroczko. « The Role of Gut Microbiota and Gut–Brain Interplay in Selected Diseases of the Central Nervous System ». International Journal of Molecular Sciences 22, no 18 (17 septembre 2021) : 10028. http://dx.doi.org/10.3390/ijms221810028.

Texte intégral
Résumé :
The gut microbiome has attracted increasing attention from researchers in recent years. The microbiota can have a specific and complex cross-talk with the host, particularly with the central nervous system (CNS), creating the so-called “gut–brain axis”. Communication between the gut, intestinal microbiota, and the brain involves the secretion of various metabolites such as short-chain fatty acids (SCFAs), structural components of bacteria, and signaling molecules. Moreover, an imbalance in the gut microbiota composition modulates the immune system and function of tissue barriers such as the blood–brain barrier (BBB). Therefore, the aim of this literature review is to describe how the gut–brain interplay may contribute to the development of various neurological disorders, combining the fields of gastroenterology and neuroscience. We present recent findings concerning the effect of the altered microbiota on neurodegeneration and neuroinflammation, including Alzheimer’s and Parkinson’s diseases, as well as multiple sclerosis. Moreover, the impact of the pathological shift in the microbiome on selected neuropsychological disorders, i.e., major depressive disorders (MDD) and autism spectrum disorder (ASD), is also discussed. Future research on the effect of balanced gut microbiota composition on the gut–brain axis would help to identify new potential opportunities for therapeutic interventions in the presented diseases.
Styles APA, Harvard, Vancouver, ISO, etc.
44

Parolini, Cinzia. « Effects of Fish n-3 PUFAs on Intestinal Microbiota and Immune System ». Marine Drugs 17, no 6 (22 juin 2019) : 374. http://dx.doi.org/10.3390/md17060374.

Texte intégral
Résumé :
Studies over several decades have documented the beneficial actions of n-3 polyunsaturated fatty acids (PUFAs), which are plentiful in fish oil, in different disease states. Mechanisms responsible for the efficacy of n-3 PUFAs include: (1) Reduction of triglyceride levels; (2) anti-arrhythmic and antithrombotic effects, and (3) resolution of inflammatory processes. The human microbiota project and subsequent studies using next-generation sequencing technology have highlighted that thousands of different microbial species are present in the human gut, and that there has been a significant variability of taxa in the microbiota composition among people. Several factors (gestational age, mode of delivery, diet, sanitation and antibiotic treatment) influence the bacterial community in the human gastrointestinal tract, and among these diet habits play a crucial role. The disturbances in the gut microbiota composition, i.e., gut dysbiosis, have been associated with diseases ranging from localized gastrointestinal disorders to neurologic, respiratory, metabolic, ocular, and cardiovascular illnesses. Many studies have been published about the effects of probiotics and prebiotics on the gut microbiota/microbioma. On the contrary, PUFAs in the gut microbiota have been less well defined. However, experimental studies suggested that gut microbiota, n-3 PUFAs, and host immune cells work together to ensure the intestinal wall integrity. This review discussed current evidence concerning the links among gut microbiota, n-3 PUFAs intake, and human inflammatory disease.
Styles APA, Harvard, Vancouver, ISO, etc.
45

Sukhina, M. A., I. A. Lyagina, A. L. Safin, S. A. Frolov et V. N. Kashnikov. « Role of intestinal microbiota in colorectal carcinogenesis (review) ». Koloproktologia 20, no 1 (18 mars 2021) : 68–76. http://dx.doi.org/10.33878/2073-7556-2021-20-1-68-76.

Texte intégral
Résumé :
The aim of the review is to show possible links between intestinal microbiota and colorectal carcinogenesis, to describe the procarcinogenic properties of microorganisms associated with the development or proliferation of colorectal cancer. The gut microbiota plays a leading role in metabolism, providing important metabolites to the macroorganism. In humans, there is a spatial variability in the qualitative and quantitative microbiota composition. The intestinal microbiota provides the colony resistance, protecting it from colonization by opportunistic and pathogenic microorganisms. There is more and more data on the role of the gut microbiota in the development of colorectal cancer. The profound study of the gut microbiome in various populations is required, which will allow to identify other microorganisms associated with the development or proliferation of colorectal cancer. It can be used as biomarkers for colorectal cancer screening and predicting the response to immunotherapy.
Styles APA, Harvard, Vancouver, ISO, etc.
46

O. Ye. Gridnyev et K. Yu. Dubrov. « Non-alcoholic fatty liver disease in the context of the gut microbiome ». Modern Gastroenterology, no 5 (31 octobre 2019) : 97–103. http://dx.doi.org/10.30978/mg-2019-5-97.

Texte intégral
Résumé :
The article presents modern views on the epidemiological situation with the non-alcoholic fatty liver disease (NAFLD) in the world, its prevalence among the population of European countries, and gender-associated characteristics. The role of the intestinal microbiome in the pathogenetic mechanisms of NAFLD development at the stage of steatosis, as well as the role of the intestinal microbiota in the progression of the disease, has been elucidated. The role of the main pathogenetic factors in the disease onset in healthy people and NAFLD development has been demonstrated, including the short chain fatty acids, bile acids, lipopolysaccharides, changes in the permeability of the intestinal wall, the balance of the main bacterial clusters. The relationship of insulin resistance with the above factors and their effects on its significance in patients with metabolic syndrome has been established. The authors showed the anatomical and functional relationships of the intestine and liver within the intestinal microbiota. The experimental data have been presented that demonstrate the effects of the intestinal microbiota on the formation and progression of non-alcoholic steatosis. The effects of intestinal microbiota metabolites on the formation of liver steatosis is separately disclosed: ethanol, bile acids, trimethylamine-N-oxide, aromatic amino acids. Moreover, the authors presented the experimental data, providing explanation for the role of the intestinal microbiota on the manifestations of the metabolic syndrome and, in particular, NAFLD. In addition, the results of clinical and preclinical studies of the effects of microbiome correction on the leading pathogenetic mechanisms of NAFLD have been shown. The effects of individual bacterial phylotypes on the course of NAFLD have been considered. The main therapeutic strategies in the treatment of the disease, based on the principles of evidence-based medicine, have been presented, as well as the trends for antibiotic therapy, probiotic therapy and fecal transplantation in the context of NAFLD therapy and their potential role.
Styles APA, Harvard, Vancouver, ISO, etc.
47

Villéger, Romain, Amélie Lopès, Guillaume Carrier, Julie Veziant, Elisabeth Billard, Nicolas Barnich, Johan Gagnière, Emilie Vazeille et Mathilde Bonnet. « Intestinal Microbiota : A Novel Target to Improve Anti-Tumor Treatment ? » International Journal of Molecular Sciences 20, no 18 (17 septembre 2019) : 4584. http://dx.doi.org/10.3390/ijms20184584.

Texte intégral
Résumé :
Recently, preclinical and clinical studies targeting several types of cancer strongly supported the key role of the gut microbiota in the modulation of host response to anti-tumoral therapies such as chemotherapy, immunotherapy, radiotherapy and even surgery. Intestinal microbiome has been shown to participate in the resistance to a wide range of anticancer treatments by direct interaction with the treatment or by indirectly stimulating host response through immunomodulation. Interestingly, these effects were described on colorectal cancer but also in other types of malignancies. In addition to their role in therapy efficacy, gut microbiota could also impact side effects induced by anticancer treatments. In the first part of this review, we summarized the role of the gut microbiome on the efficacy and side effects of various anticancer treatments and underlying mechanisms. In the second part, we described the new microbiota-targeting strategies, such as probiotics and prebiotics, antibiotics, fecal microbiota transplantation and physical activity, which could be effective adjuvant therapies developed in order to improve anticancer therapeutic efficiency.
Styles APA, Harvard, Vancouver, ISO, etc.
48

Gérard, Philippe. « The crosstalk between the gut microbiota and lipids ». OCL 27 (2020) : 70. http://dx.doi.org/10.1051/ocl/2020070.

Texte intégral
Résumé :
The human intestine harbours a complex and diverse bacterial community called the gut microbiota. This microbiota, stable during the lifetime, is specific of each individual despite the existence of a phylogenetic core shared by the majority of adults. The influence of the gut microbiota on host’s physiology has been largely studied using germfree animals and studies using these animal models have revealed that the effects of lipids on host physiology are microbiota-dependent. Studies in mice have also shown that a high-fat diet rapidly and reproducibly alters the gut microbiome. In humans, dietary fat interventions did not lead to strong and consistent modifications of the microbiota composition. Nevertheless, an association between total fat intake and the reduction of the microbiota richness has been repeatedly found. Interestingly, different types of fat exert different or even opposite effects on the microbiota. Concurrently, the gut microbiota is able to convert the lipids entering the colon, including fatty acids or cholesterol, leading to the production of metabolites with potential health effects.
Styles APA, Harvard, Vancouver, ISO, etc.
49

Forkosh, Esther, et Yaron Ilan. « The heart-gut axis : new target for atherosclerosis and congestive heart failure therapy ». Open Heart 6, no 1 (avril 2019) : e000993. http://dx.doi.org/10.1136/openhrt-2018-000993.

Texte intégral
Résumé :
The human gut microbiota has been identified as a possible novel risk factor for cardiovascular disease. The intestinal microbiome plays a role in the pathogenesis of atherosclerosis and heart failure. Even though studies in rodents suggested that gut microbes may affect the risk of heart disease, this link has not been shown in humans. In the present study, we review several potential mechanisms by which the gut microbiome and bacterial translocation are associated with the development of cardiac disorders making them potential targets for novel therapeutic measures for these conditions. Modulation of the gut microbiota as a mechanism for altering the pathogenesis of disorders is an area of growing interest. Alteration in the gut microbiota is being explored as a method of reducing risk factors associated with cardiac diseases.
Styles APA, Harvard, Vancouver, ISO, etc.
50

Deng, Lei, Lukasz Wojciech, Nicholas R. J. Gascoigne, Guangneng Peng et Kevin S. W. Tan. « New insights into the interactions between Blastocystis, the gut microbiota, and host immunity ». PLOS Pathogens 17, no 2 (25 février 2021) : e1009253. http://dx.doi.org/10.1371/journal.ppat.1009253.

Texte intégral
Résumé :
The human gut microbiota is a diverse and complex ecosystem that is involved in beneficial physiological functions as well as disease pathogenesis. Blastocystis is a common protistan parasite and is increasingly recognized as an important component of the gut microbiota. The correlations between Blastocystis and other communities of intestinal microbiota have been investigated, and, to a lesser extent, the role of this parasite in maintaining the host immunological homeostasis. Despite recent studies suggesting that Blastocystis decreases the abundance of beneficial bacteria, most reports indicate that Blastocystis is a common component of the healthy gut microbiome. This review covers recent finding on the potential interactions between Blastocystis and the gut microbiota communities and its roles in regulating host immune responses.
Styles APA, Harvard, Vancouver, ISO, etc.
Nous offrons des réductions sur tous les plans premium pour les auteurs dont les œuvres sont incluses dans des sélections littéraires thématiques. Contactez-nous pour obtenir un code promo unique!

Vers la bibliographie