Journal articles on the topic 'Host-microbiome interaction'

To see the other types of publications on this topic, follow the link: Host-microbiome interaction.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Host-microbiome interaction.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Kelly, Paul. "Nutrition, intestinal defence and the microbiome." Proceedings of the Nutrition Society 69, no. 2 (March 5, 2010): 261–68. http://dx.doi.org/10.1017/s0029665110000108.

Full text
Abstract:
The interaction between nutrition and infection was the subject of important work by several groups in the 1960s. The explosion of knowledge in immunology, including innate immunity, has led to increased understanding of the impact of nutrition on host defence, but much more work needs to be done in this area. In the last decade an increasing volume of work has opened up the previously obscure world of human endogenous flora. This work suggests that the microbiome, the total genetic pool of the microbiota, contributes to the already complex interaction between nutrition and infectious disease. The established concept that nutritional status, host defence and infection all impact on each other now has to be expanded into a multiple interaction, with the microbiota interacting with all three other elements. There is good evidence that the microbiome programmes host defence and drives a metabolome that impacts on energy balance, and indeed on some micronutrients. In turn, host defence shapes the microbiome, and nutritional status, particularly micronutrient status, helps determine several elements of host defence. While interventions in this area are in their infancy, the understanding of interactions that already have an enormous impact on global health is now at a threshold. The present review explores the evidence for these interactions with a view to putting potential interventions into the context of a conceptual framework.
APA, Harvard, Vancouver, ISO, and other styles
2

Loskutov, S. I., S. N. Proshin, and D. S. Ryabukhin. "Evolutionary aspects of gastrointestinal tract microbiome-host interaction underlying gastrointestinal barrier integrity." Russian Journal of Infection and Immunity 12, no. 5 (November 16, 2022): 819–26. http://dx.doi.org/10.15789/2220-7619-eao-1633.

Full text
Abstract:
In the host sustenance and homeostasis, the microbiome is a key component in the functional system. Throughout ontogenetic development, microbiome including that of the gastrointestinal tract (GIT) is the vital factor that ensures not only host functioning, but also its interaction with environment. To uncover the mechanisms underlying GIT microbiome showing a decisive influence on host organism, a systematic approach is needed, because diverse microorganisms are predominantly localized in different parts of the GIT. Recently, a new interdisciplinary direction of science, nanobioinformatics that has been extensively developed considers gene networks as the major object of study representing a coordinated group of genes that functionally account for formation and phenotypic disclosure of various host traits. Here, an important place should be provided to the genetically determined level of the gastrointestinal tract microbiome, its interaction at the level of the host food systems. There have been increasing evidence indicating that the microbiome is directly involved in the pathogenesis of host diseases showing a multi-layered interaction with host metabolic and immune systems. At the same time, the microbial community is unevenly distributed throughout the gastrointestinal tract, and its different portions are variously active while interacting with the host immune system. The architecture of interaction between the microbiome and host cells is extremely complex, and the interaction of individual cells, at the same time, varies greatly. Bacteria colonizing the crypts of the small intestine regulate enterocyte proliferation by affecting DNA replication and gene expression, while bacteria at the tip of the intestinal villi mediate gene expression responsible for metabolism and immune response. Enterocytes and Paneth cells, in turn, regulate the vital activity of the community of microorganisms through the production of polysaccharides (carbohydrates) and antibacterial factors on their surface. Thus, the integrity of the gastrointestinal barrier (GIB) is maintained, which protects the body from infections and inflammation, while violation of its integrity leads to a number of diseases. It has been shown that depending on the dominance of certain types of bacteria the microbiome can maintain or disrupt GIB integrity. The structural and functional GIB integrity is important for body homeostasis. To date, at least 50 proteins have been characterized as being involved in the structural and functional integrability of tight junctions between gastrointestinal tract epithelial cells. The current review comprehensively discusses such issues and presents original research carried out at various facilities of translational biomedicine.
APA, Harvard, Vancouver, ISO, and other styles
3

Weersma, Rinse K., Alexandra Zhernakova, and Jingyuan Fu. "Interaction between drugs and the gut microbiome." Gut 69, no. 8 (May 14, 2020): 1510–19. http://dx.doi.org/10.1136/gutjnl-2019-320204.

Full text
Abstract:
The human gut microbiome is a complex ecosystem that can mediate the interaction of the human host with their environment. The interaction between gut microbes and commonly used non-antibiotic drugs is complex and bidirectional: gut microbiome composition can be influenced by drugs, but, vice versa, the gut microbiome can also influence an individual’s response to a drug by enzymatically transforming the drug’s structure and altering its bioavailability, bioactivity or toxicity (pharmacomicrobiomics). The gut microbiome can also indirectly impact an individual’s response to immunotherapy in cancer treatment. In this review we discuss the bidirectional interactions between microbes and drugs, describe the changes in gut microbiota induced by commonly used non-antibiotic drugs, and their potential clinical consequences and summarise how the microbiome impacts drug effectiveness and its role in immunotherapy. Understanding how the microbiome metabolises drugs and reduces treatment efficacy will unlock the possibility of modulating the gut microbiome to improve treatment.
APA, Harvard, Vancouver, ISO, and other styles
4

Li, Yue-Han, Yuan-You Yang, Zhi-Gang Wang, and Zhuo Chen. "Emerging Function of Ecotype-Specific Splicing in the Recruitment of Commensal Microbiome." International Journal of Molecular Sciences 23, no. 9 (April 27, 2022): 4860. http://dx.doi.org/10.3390/ijms23094860.

Full text
Abstract:
In recent years, host–microbiome interactions in both animals and plants has emerged as a novel research area for studying the relationship between host organisms and their commensal microbial communities. The fitness advantages of this mutualistic interaction can be found in both plant hosts and their associated microbiome, however, the driving forces mediating this beneficial interaction are poorly understood. Alternative splicing (AS), a pivotal post-transcriptional mechanism, has been demonstrated to play a crucial role in plant development and stress responses among diverse plant ecotypes. This natural variation of plants also has an impact on their commensal microbiome. In this article, we review the current progress of plant natural variation on their microbiome community, and discuss knowledge gaps between AS regulation of plants in response to their intimately related microbiota. Through the impact of this article, an avenue could be established to study the biological mechanism of naturally varied splicing isoforms on plant-associated microbiome assembly.
APA, Harvard, Vancouver, ISO, and other styles
5

Taschuk, Ryan, and Philip J. Griebel. "Commensal microbiome effects on mucosal immune system development in the ruminant gastrointestinal tract." Animal Health Research Reviews 13, no. 1 (June 2012): 129–41. http://dx.doi.org/10.1017/s1466252312000096.

Full text
Abstract:
AbstractCommensal microflora play many roles within the mammalian gastrointestinal tract (GIT) that benefit host physiology by way of direct or indirect interactions with mucosal surfaces. Commensal flora comprises members across all microbial phyla, although predominantly bacterial, with population dynamics varying with host species, genotype, and environmental factors. Little is known, however, about the complex mechanisms regulating host–commensal interactions that underlie this mutually beneficial relationship and how alterations in the microbiome may influence host development and susceptibility to infection. Research into the gut microbiome has intensified as it becomes increasingly evident that symbiont–host interactions have a significant impact on mucosal immunity and health. Furthermore, evidence that microbial populations vary significantly throughout the GIT suggest that regional differences in the microbiome may also influence immune function within distinct compartments of the GIT. Postpartum colonization of the GIT has been shown to have a direct effect on mucosal immune system development, but information is limited regarding regional effects of the microbiome on the development, activation, and maturation of the mucosal immune system. This review discusses factors influencing the colonization and establishment of the microbiome throughout the GIT of newborn calves and the evidence that regional differences in the microbiome influence mucosal immune system development and maturation. The implications of this complex interaction are also discussed in terms of possible effects on responses to enteric pathogens and vaccines.
APA, Harvard, Vancouver, ISO, and other styles
6

Karaduta, Oleg, Zeljko Dvanajscak, and Boris Zybailov. "Metaproteomics—An Advantageous Option in Studies of Host-Microbiota Interaction." Microorganisms 9, no. 5 (April 30, 2021): 980. http://dx.doi.org/10.3390/microorganisms9050980.

Full text
Abstract:
Gut microbiome contributes to host health by maintaining homeostasis, increasing digestive efficiency, and facilitating the development of the immune system. Manipulating gut microbiota is being recognized as a therapeutic target to manage various chronic diseases. The therapeutic manipulation of the intestinal microbiome is achieved through diet modification, the administration of prebiotics, probiotics, or antibiotics, and more recently, fecal microbiome transplantation (FMT). In this opinion paper, we give a perspective on the current status of application of multi-omics technologies in the analysis of host-microbiota interactions. The aim of this paper was to highlight the strengths of metaproteomics, which integrates with and often relies on other approaches.
APA, Harvard, Vancouver, ISO, and other styles
7

Brinker, Pina, Michael C. Fontaine, Leo W. Beukeboom, and Joana Falcao Salles. "Host, Symbionts, and the Microbiome: The Missing Tripartite Interaction." Trends in Microbiology 27, no. 6 (June 2019): 480–88. http://dx.doi.org/10.1016/j.tim.2019.02.002.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Chimenti, Maria Sole, Carlo Perricone, Lucia Novelli, Francesco Caso, Luisa Costa, Dimitrios Bogdanos, Paola Conigliaro, et al. "Interaction between microbiome and host genetics in psoriatic arthritis." Autoimmunity Reviews 17, no. 3 (March 2018): 276–83. http://dx.doi.org/10.1016/j.autrev.2018.01.002.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Pinker, Elisha, and Timur Tuganbaev. "Microbiome Composition and Circadian Rhythm Disruption Alters Epithelial Barrier Integrity." Columbia Undergraduate Science Journal 15 (May 24, 2021): 6–15. http://dx.doi.org/10.52214/cusj.v15i1.7408.

Full text
Abstract:
The intestine is home to one of the most complex ecological communities, termed the human gut microbiome. The gut microbiome modulates a wide range of human diseases from diabetes to neurological disorders to cancer. Separating the host and the gut microbiome is the epithelial barrier. The intestinal epithelium serves as an adaptive interaction hub between the host and microbiome that plays an important role in deciding the outcome of host-microbiome interactions. Regulation of epithelial barrier permeability to ions, nutrients and microbiome metabolites is known to be a tightly controlled process on the host side. However, whether the microbiome community also affects epithelial permeability remains unclear. Here, we show that alterations in microbiota composition by treatment with antibiotics selectively targeting specific members of the microbiome community impacts the permeability of the intestine. Additionally, modulating the microbiome through other methods such as altering diet composition shows changes in permeability of the epithelial barrier. As daily feeding rhythm entrains diurnal fluctuations in microbiome, we have set out to measure epithelial barrier permeability throw out the clock. We have discovered that the permeability of the intestinal epithelial barrier exhibits circadian rhythms in mice. Disruption of these rhythms, through jet-lag or genetic deficiencies in circadian machinery, consequently alters epithelial barrier integrity. Together, these findings provide evidence that disruptions in circadian rhythms as well as alterations in microbiome composition have direct consequences in intestinal permeability, and that microbiome might serve as a tool in regulating epithelium permeability.
APA, Harvard, Vancouver, ISO, and other styles
10

Shine, Emilee E., and Jason M. Crawford. "Molecules from the Microbiome." Annual Review of Biochemistry 90, no. 1 (June 20, 2021): 789–815. http://dx.doi.org/10.1146/annurev-biochem-080320-115307.

Full text
Abstract:
The human microbiome encodes a second genome that dwarfs the genetic capacity of the host. Microbiota-derived small molecules can directly target human cells and their receptors or indirectly modulate host responses through functional interactions with other microbes in their ecological niche. Their biochemical complexity has profound implications for nutrition, immune system development, disease progression, and drug metabolism, as well as the variation in these processes that exists between individuals. While the species composition of the human microbiome has been deeply explored, detailed mechanistic studies linking specific microbial molecules to host phenotypes are still nascent. In this review, we discuss challenges in decoding these interaction networks, which require interdisciplinary approaches that combine chemical biology, microbiology, immunology, genetics, analytical chemistry, bioinformatics, and synthetic biology. We highlight important classes of microbiota-derived small molecules and notable examples. An understanding of these molecular mechanisms is central to realizing the potential of precision microbiome editing in health, disease, and therapeutic responses.
APA, Harvard, Vancouver, ISO, and other styles
11

Balakrishnan, Kalpana, Divya Sivanesan, Gaanappriya Mohan, Sachin Gunthe, and Rama Verma. "Importance of Interkingdom Interactions Among Oral Microbiome Towards Caries Development – A Review." Journal of Immunological Sciences 5, no. 2 (May 30, 2021): 27–35. http://dx.doi.org/10.29245/2578-3009/2021/2.1211.

Full text
Abstract:
The human microbiome plays a crucial role in health and disease conditions. These microbiomes constitute a structured, coordinated microbial network throughout the human body. The oral cavity harbors one of the extensively diverse bacteria in the human system. Although many studies emphasize bacteriome and its interaction with the host system, very little attention is given to candidate phyla radiation (CPR), fungal components, and its interkingdom interaction in the oral microecology even with advanced techniques. The interkingdom interactions among caries causing microbes trigger the pathogenesis of bacterial diseases and cause ecological shifts and affect the host system. Studying the complex relations among the diverse oral microbiome and its host, especially CPR phyla and fungi, would give a holistic view of the caries etiology. This review provides evidence on the interkingdom interaction that establishes a complex community that could help predict future oral and systemic diseases.
APA, Harvard, Vancouver, ISO, and other styles
12

Tsolis, Renée M., and Andreas J. Bäumler. "Gastrointestinal host-pathogen interaction in the age of microbiome research." Current Opinion in Microbiology 53 (February 2020): 78–89. http://dx.doi.org/10.1016/j.mib.2020.03.002.

Full text
APA, Harvard, Vancouver, ISO, and other styles
13

Moeller, Andrew H., Steffen Foerster, Michael L. Wilson, Anne E. Pusey, Beatrice H. Hahn, and Howard Ochman. "Social behavior shapes the chimpanzee pan-microbiome." Science Advances 2, no. 1 (January 2016): e1500997. http://dx.doi.org/10.1126/sciadv.1500997.

Full text
Abstract:
Animal sociality facilitates the transmission of pathogenic microorganisms among hosts, but the extent to which sociality enables animals’ beneficial microbial associations is poorly understood. The question is critical because microbial communities, particularly those in the gut, are key regulators of host health. We show evidence that chimpanzee social interactions propagate microbial diversity in the gut microbiome both within and between host generations. Frequent social interaction promotes species richness within individual microbiomes as well as homogeneity among the gut community memberships of different chimpanzees. Sampling successive generations across multiple chimpanzee families suggests that infants inherited gut microorganisms primarily through social transmission. These results indicate that social behavior generates a pan-microbiome, preserving microbial diversity across evolutionary time scales and contributing to the evolution of host species–specific gut microbial communities.
APA, Harvard, Vancouver, ISO, and other styles
14

Maglione, Alessandro, Miriam Zuccalà, Martina Tosi, Marinella Clerico, and Simona Rolla. "Host Genetics and Gut Microbiome: Perspectives for Multiple Sclerosis." Genes 12, no. 8 (July 29, 2021): 1181. http://dx.doi.org/10.3390/genes12081181.

Full text
Abstract:
As a complex disease, Multiple Sclerosis (MS)’s etiology is determined by both genetic and environmental factors. In the last decade, the gut microbiome has emerged as an important environmental factor, but its interaction with host genetics is still unknown. In this review, we focus on these dual aspects of MS pathogenesis: we describe the current knowledge on genetic factors related to MS, based on genome-wide association studies, and then illustrate the interactions between the immune system, gut microbiome and central nervous system in MS, summarizing the evidence available from Experimental Autoimmune Encephalomyelitis mouse models and studies in patients. Finally, as the understanding of influence of host genetics on the gut microbiome composition in MS is in its infancy, we explore this issue based on the evidence currently available from other autoimmune diseases that share with MS the interplay of genetic with environmental factors (Inflammatory Bowel Disease, Rheumatoid Arthritis and Systemic Lupus Erythematosus), and discuss avenues for future research.
APA, Harvard, Vancouver, ISO, and other styles
15

Zeng, Yao, and Qiaoyi Liang. "Nasal Microbiome and Its Interaction with the Host in Childhood Asthma." Cells 11, no. 19 (October 7, 2022): 3155. http://dx.doi.org/10.3390/cells11193155.

Full text
Abstract:
Childhood asthma is a major chronic non-communicable disease in infants and children, often triggered by respiratory tract infections. The nasal cavity is a reservoir for a broad variety of commensal microbes and potential pathogens associated with respiratory illnesses including asthma. A healthy nasal microenvironment has protective effects against respiratory tract infections. The first microbial colonisation in the nasal region is initiated immediately after birth. Subsequently, colonisation by nasal microbiota during infancy plays important roles in rapidly establishing immune homeostasis and the development and maturation of the immune system. Dysbiosis of microbiota residing in the mucosal surfaces, such as the nasopharynx and guts, triggers immune modulation, severe infection, and exacerbation events. Nasal microbiome dysbiosis is related to the onset of symptomatic infections. Dynamic interactions between viral infections and the nasal microbiota in early life affect the later development of respiratory infections. In this review, we summarise the existing findings related to nasal microbiota colonisation, dynamic variations, and host–microbiome interactions in childhood health and respiratory illness with a particular examination of asthma. We also discuss our current understanding of biases produced by environmental factors and technical concerns, the importance of standardised research methods, and microbiome modification for the prevention or treatment of childhood asthma. This review lays the groundwork for paying attention to an essential but less emphasized topic and improves the understanding of the overall composition, dynamic changes, and influence of the nasal microbiome associated with childhood asthma.
APA, Harvard, Vancouver, ISO, and other styles
16

Houwenhuyse, Shira, Robby Stoks, Shinjini Mukherjee, and Ellen Decaestecker. "Locally adapted gut microbiomes mediate host stress tolerance." ISME Journal 15, no. 8 (March 3, 2021): 2401–14. http://dx.doi.org/10.1038/s41396-021-00940-y.

Full text
Abstract:
AbstractWhile evidence for the role of the microbiome in shaping host stress tolerance is becoming well-established, to what extent this depends on the interaction between the host and its local microbiome is less clear. Therefore, we investigated whether locally adapted gut microbiomes affect host stress tolerance. In the water flea Daphnia magna, we studied if the host performs better when receiving a microbiome from their source region than from another region when facing a stressful condition, more in particular exposure to the toxic cyanobacteria Microcystis aeruginosa. Therefore, a reciprocal transplant experiment was performed in which recipient, germ-free D. magna, isolated from different ponds, received a donor microbiome from sympatric or allopatric D. magna that were pre-exposed to toxic cyanobacteria or not. We tested for effects on host life history traits and gut microbiome composition. Our data indicate that Daphnia interact with particular microbial strains mediating local adaptation in host stress tolerance. Most recipient D. magna individuals performed better when inoculated with sympatric than with allopatric microbiomes. This effect was most pronounced when the donors were pre-exposed to the toxic cyanobacteria, but this effect was also pond and genotype dependent. We discuss how this host fitness benefit is associated with microbiome diversity patterns.
APA, Harvard, Vancouver, ISO, and other styles
17

Ariyoshi, Tadashi, Mao Hagihara, Susumu Tomono, Shuhei Eguchi, Ayaka Minemura, Daiki Miura, Kentaro Oka, Motomichi Takahashi, Yuka Yamagishi, and Hiroshige Mikamo. "Clostridium butyricum MIYAIRI 588 Modifies Bacterial Composition under Antibiotic-Induced Dysbiosis for the Activation of Interactions via Lipid Metabolism between the Gut Microbiome and the Host." Biomedicines 9, no. 8 (August 22, 2021): 1065. http://dx.doi.org/10.3390/biomedicines9081065.

Full text
Abstract:
The gut microbiome is closely related to gut metabolic functions, and the gut microbiome and host metabolic functions affect each other. Clostridium butyricum MIYAIRI 588 (CBM 588) upregulates protectin D1 production in host colon tissue following G protein-coupled receptor (GPR) 120 activation to protect gut epithelial cells under antibiotic-induced dysbiosis. However, how CBM 588 enhances polyunsaturated fatty acid (PUFA) metabolites remains unclear. Therefore, we focused on the metabolic function alterations of the gut microbiome after CBM 588 and protectin D1 administration to reveal the interaction between the host and gut microbiome through lipid metabolism during antibiotic-induced dysbiosis. Consequently, CBM 588 modified gut microbiome and increased the butyric acid and oleic acid content. These lipid metabolic modifications induced GPR activation, which is a trigger of ERK 1/2 signaling and directed differentiation of downstream immune cells in the host colon tissue. Moreover, endogenous protectin D1 modified the gut microbiome, similar to CBM 588. This is the first study to report that CBM 588 influences the interrelationship between colon tissue and the gut microbiome through lipid metabolism. These findings provide insights into the mechanisms of prevention and recovery from inflammation and the improvement of host metabolism by CBM 588.
APA, Harvard, Vancouver, ISO, and other styles
18

Abdul Rahman, Nur Sabrina Natasha, Nur Wahida Abdul Hamid, and Kalaivani Nadarajah. "Effects of Abiotic Stress on Soil Microbiome." International Journal of Molecular Sciences 22, no. 16 (August 21, 2021): 9036. http://dx.doi.org/10.3390/ijms22169036.

Full text
Abstract:
Rhizospheric organisms have a unique manner of existence since many factors can influence the shape of the microbiome. As we all know, harnessing the interaction between soil microbes and plants is critical for sustainable agriculture and ecosystems. We can achieve sustainable agricultural practice by incorporating plant-microbiome interaction as a positive technology. The contribution of this interaction has piqued the interest of experts, who plan to do more research using beneficial microorganism in order to accomplish this vision. Plants engage in a wide range of interrelationship with soil microorganism, spanning the entire spectrum of ecological potential which can be mutualistic, commensal, neutral, exploitative, or competitive. Mutualistic microorganism found in plant-associated microbial communities assist their host in a number of ways. Many studies have demonstrated that the soil microbiome may provide significant advantages to the host plant. However, various soil conditions (pH, temperature, oxygen, physics-chemistry and moisture), soil environments (drought, submergence, metal toxicity and salinity), plant types/genotype, and agricultural practices may result in distinct microbial composition and characteristics, as well as its mechanism to promote plant development and defence against all these stressors. In this paper, we provide an in-depth overview of how the above factors are able to affect the soil microbial structure and communities and change above and below ground interactions. Future prospects will also be discussed.
APA, Harvard, Vancouver, ISO, and other styles
19

Dohlman, Anders B., and Xiling Shen. "Mapping the microbial interactome: Statistical and experimental approaches for microbiome network inference." Experimental Biology and Medicine 244, no. 6 (March 16, 2019): 445–58. http://dx.doi.org/10.1177/1535370219836771.

Full text
Abstract:
Advances in high-throughput sequencing have ushered in a new era of research into the gut microbiome and its role in human health and disease. However, due to the unique characteristics of microbiome survey data, their use for the detection of ecological interaction networks remains a considerable challenge, and a field of active methodological development. In this review, we discuss the landscape of existing statistical and experimental methods for detecting and characterizing microbial interactions, as well as the role that host and environmental metabolic signals play in mediating the behavior of these networks. Numerous statistical tools for microbiome network inference have been developed. Yet due to tool-specific biases, the networks identified by these methods are often discordant, motivating a need for the development of more general tools, the use of ensemble approaches, and the incorporation of prior knowledge into prediction. By elucidating the complex dynamics of the microbial interactome, we will enhance our understanding of the microbiome’s role in disease, more precisely predict the microbiome’s response to perturbation, and inform the development of future therapeutic strategies for microbiome-related disease. Impact statement This review provides a comprehensive description of experimental and statistical tools used for network analyses of the human gut microbiome. Understanding the system dynamics of microbial interactions may lead to the improvement of therapeutic approaches for managing microbiome-associated diseases. Microbiome network inference tools have been developed and applied to both cross-sectional and longitudinal experimental designs, as well as to multi-omic datasets, with the goal of untangling the complex web of microbe-host, microbe-environmental, and metabolism-mediated microbial interactions. The characterization of these interaction networks may lead to a better understanding of the systems dynamics of the human gut microbiome, augmenting our knowledge of the microbiome’s role in human health, and guiding the optimization of effective, precise, and rational therapeutic strategies for managing microbiome-associated disease.
APA, Harvard, Vancouver, ISO, and other styles
20

Kononova, Svetlana, Ekaterina Litvinova, Timur Vakhitov, Maria Skalinskaya, and Stanislav Sitkin. "Acceptive Immunity: The Role of Fucosylated Glycans in Human Host–Microbiome Interactions." International Journal of Molecular Sciences 22, no. 8 (April 8, 2021): 3854. http://dx.doi.org/10.3390/ijms22083854.

Full text
Abstract:
The growth in the number of chronic non-communicable diseases in the second half of the past century and in the first two decades of the new century is largely due to the disruption of the relationship between the human body and its symbiotic microbiota, and not pathogens. The interaction of the human immune system with symbionts is not accompanied by inflammation, but is a physiological norm. This is achieved via microbiota control by the immune system through a complex balance of pro-inflammatory and suppressive responses, and only a disturbance of this balance can trigger pathophysiological mechanisms. This review discusses the establishment of homeostatic relationships during immune system development and intestinal bacterial colonization through the interaction of milk glycans, mucins, and secretory immunoglobulins. In particular, the role of fucose and fucosylated glycans in the mechanism of interactions between host epithelial and immune cells is discussed.
APA, Harvard, Vancouver, ISO, and other styles
21

Grifka-Walk, Heather M., Nicholas V. Pinkham, Narayanaganesh Balasubramanian, Brittany R. Jenkins, Nicholas Looby, Benjamin Deuling, Steve D. Swain, Hailey Liss, Seth T. Walk, and Douglas J. Kominsky. "Discovery of a protective, transmissible, and type 2 inflammation-skewing intestinal microbiome." Journal of Immunology 204, no. 1_Supplement (May 1, 2020): 83.19. http://dx.doi.org/10.4049/jimmunol.204.supp.83.19.

Full text
Abstract:
Abstract We recently discovered a transmissible, dominant, and remarkably protective intestinal microbiome (a “Magical Microbiome,” MM) in the setting of a murine model of colitis. The objective of this project is to fill current knowledge gaps regarding the molecular mechanisms underlying protection in this novel model of microbiome-mediated protection. Protection can be transmitted to unmanipulated conventional or germ-free C57Bl/6 mice via cohousing or oral gavage of MM+ stool. During DSS-induced colitis, MM+ mice develop less weight loss, intestinal pathology, and production of proinflammatory molecules relative to genetically identical C57Bl/6 MM-controls. Preliminary results suggest that the eukaryotic and bacterial microbiome under investigation re-programs the host inflammation to prevent disease or promote repair. MM-mediated protection is IL-13-dependent and associated with enhanced type 2 inflammation, as transcript levels of type 2-associated genes including IL-4, IL-13, and Fizz1 were significantly higher in colons of MM+ mice. 16s rRNA gene sequencing and eukaryote screening demonstrated that MM+ mice have a bacterial microbiome distinct from controls and host a novel Tritrichomonas species. Transfer of Tritrichomonas to wild-type mice correlated with protection, but vancomycin treatment ameliorates the protection while maintaining Tritrichomonas, suggesting a possible interaction between bacteria and protist. Stool metabolites are also distinct between MM+ and MM− hosts, further supporting a distinct microbiome function. Overall, we possess a unique microbiome that can be used to understand protective host-microbe interactions and identify novel therapeutic targets for inflammatory diseases.
APA, Harvard, Vancouver, ISO, and other styles
22

Michielan, Andrea. "Host-microbiome interaction in Crohn’s disease: A familiar or familial issue?" World Journal of Gastrointestinal Pathophysiology 6, no. 4 (2015): 159. http://dx.doi.org/10.4291/wjgp.v6.i4.159.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Pan, Deng, and Zhongtang Yu. "Intestinal microbiome of poultry and its interaction with host and diet." Gut Microbes 5, no. 1 (October 31, 2013): 108–19. http://dx.doi.org/10.4161/gmic.26945.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Gong, Sanqiang, Xuejie Jin, Lijuan Ren, Yehui Tan, and Xiaomin Xia. "Unraveling Heterogeneity of Coral Microbiome Assemblages in Tropical and Subtropical Corals in the South China Sea." Microorganisms 8, no. 4 (April 21, 2020): 604. http://dx.doi.org/10.3390/microorganisms8040604.

Full text
Abstract:
Understanding the coral microbiome is critical for predicting the fidelity of coral symbiosis with growing surface seawater temperature (SST). However, how the coral microbiome will respond to increasing SST is still understudied. Here, we compared the coral microbiome assemblages among 73 samples across six typical South China Sea coral species in two thermal regimes. The results revealed that the composition of microbiome varied across both coral species and thermal regimes, except for Porites lutea. The tropical coral microbiome displayed stronger heterogeneity and had a more un-compacted ecological network than subtropical coral microbiome. The coral microbiome was more strongly determined by environmental factors than host specificity. γ- (32%) and α-proteobacteria (19%), Bacteroidetes (14%), Firmicutes (14%), Actinobacteria (6%) and Cyanobacteria (2%) dominated the coral microbiome. Additionally, bacteria inferred to play potential roles in host nutrients metabolism, several keystone bacteria detected in human and plant rhizospheric microbiome were retrieved in explored corals. This study not only disentangles how different host taxa and microbiome interact and how such an interaction is affected by thermal regimes, but also identifies previously unrecognized keystone bacteria in corals, and also infers the community structure of coral microbiome will be changed from a compacted to an un-compacted network under elevated SST.
APA, Harvard, Vancouver, ISO, and other styles
25

Zhang, Ping. "Influence of Foods and Nutrition on the Gut Microbiome and Implications for Intestinal Health." International Journal of Molecular Sciences 23, no. 17 (August 24, 2022): 9588. http://dx.doi.org/10.3390/ijms23179588.

Full text
Abstract:
Food components in our diet provide not only necessary nutrients to our body but also substrates for the mutualistic microbial flora in our gastrointestinal tract, termed the gut microbiome. Undigested food components are metabolized to a diverse array of metabolites. Thus, what we eat shapes the structure, composition, and function of the gut microbiome, which interacts with the gut epithelium and mucosal immune system and maintains intestinal homeostasis in a healthy state. Alterations of the gut microbiome are implicated in many diseases, such as inflammatory bowel disease (IBD). There is growing interest in nutritional therapy to target the gut microbiome in IBD. Investigations into dietary effects on the composition changes in the gut microbiome flourished in recent years, but few focused on gut physiology. This review summarizes the current knowledge regarding the impacts of major food components and their metabolites on the gut and health consequences, specifically within the GI tract. Additionally, the influence of the diet on the gut microbiome-host immune system interaction in IBD is also discussed. Understanding the influence of the diet on the interaction of the gut microbiome and the host immune system will be useful in developing nutritional strategies to maintain gut health and restore a healthy microbiome in IBD.
APA, Harvard, Vancouver, ISO, and other styles
26

Avery, Ellen G., Hendrik Bartolomaeus, Andras Maifeld, Lajos Marko, Helge Wiig, Nicola Wilck, Stephan P. Rosshart, Sofia K. Forslund, and Dominik N. Müller. "The Gut Microbiome in Hypertension." Circulation Research 128, no. 7 (April 2, 2021): 934–50. http://dx.doi.org/10.1161/circresaha.121.318065.

Full text
Abstract:
The pathogenesis of hypertension is known to involve a diverse range of contributing factors including genetic, environmental, hormonal, hemodynamic and inflammatory forces, to name a few. There is mounting evidence to suggest that the gut microbiome plays an important role in the development and pathogenesis of hypertension. The gastrointestinal tract, which houses the largest compartment of immune cells in the body, represents the intersection of the environment and the host. Accordingly, lifestyle factors shape and are modulated by the microbiome, modifying the risk for hypertensive disease. One well-studied example is the consumption of dietary fibers, which leads to the production of short-chain fatty acids and can contribute to the expansion of anti-inflammatory immune cells, consequently protecting against the progression of hypertension. Dietary interventions such as fasting have also been shown to impact hypertension via the microbiome. Studying the microbiome in hypertensive disease presents a variety of unique challenges to the use of traditional model systems. Integrating microbiome considerations into preclinical research is crucial, and novel strategies to account for reciprocal host-microbiome interactions, such as the wildling mouse model, may provide new opportunities for translation. The intricacies of the role of the microbiome in hypertensive disease is a matter of ongoing research, and there are several technical considerations which should be accounted for moving forward. In this review we provide insights into the host-microbiome interaction and summarize the evidence of its importance in the regulation of blood pressure. Additionally, we provide recommendations for ongoing and future research, such that important insights from the microbiome field at large can be readily integrated in the context of hypertension.
APA, Harvard, Vancouver, ISO, and other styles
27

GLENDINNING, LAURA, NORMAN NAUSCH, ANDREW FREE, DAVID W. TAYLOR, and FRANCISCA MUTAPI. "The microbiota and helminths: sharing the same niche in the human host." Parasitology 141, no. 10 (June 5, 2014): 1255–71. http://dx.doi.org/10.1017/s0031182014000699.

Full text
Abstract:
SUMMARYHuman gastrointestinal bacteria often share their environment with parasitic worms, allowing physical and physiological interaction between the two groups. Such associations have the potential to affect host health as well as the bacterial and helminth populations. Although still in its early stages, research on the interaction between the microbiome and parasitic helminths in humans offers the potential to improve health by manipulating the microbiome. Previously, supplementation with various nutritional compounds has been found to increase the abundance of potentially beneficial gut commensal bacteria. Thus, nutritional microbiome manipulation to produce an environment which may decrease malnutrition associated with helminth infection and/or aid host recovery from disease is conceivable. This review discusses the influence of the gut microbiota and helminths on host nutrition and immunity and the subsequent effects on the human host's overall health. It also discusses changes occurring in the microbiota upon helminth infections and the underlying mechanisms leading to these changes. There are still significant knowledge gaps which need to be filled before meaningful progress can be made in translating knowledge from studying the human gut microbiome into therapeutic strategies. Ultimately this review aims to discuss our current knowledge as well as highlight areas requiring further investigation.
APA, Harvard, Vancouver, ISO, and other styles
28

Bulteel, Lore, Shira Houwenhuyse, Steven A. J. Declerck, and Ellen Decaestecker. "The Role of Microbiome and Genotype in Daphnia magna upon Parasite Re-Exposure." Genes 12, no. 1 (January 7, 2021): 70. http://dx.doi.org/10.3390/genes12010070.

Full text
Abstract:
Recently, it has been shown that the community of gut microorganisms plays a crucial role in host performance with respect to parasite tolerance. Knowledge, however, is lacking on the role of the gut microbiome in mediating host tolerance after parasite re-exposure, especially considering multiple parasite infections. We here aimed to fill this knowledge gap by studying the role of the gut microbiome on tolerance in Daphnia magna upon multiple parasite species re-exposure. Additionally, we investigated the role of the host genotype in the interaction between the gut microbiome and the host phenotypic performance. A microbiome transplant experiment was performed in which three germ-free D. magna genotypes were exposed to a gut microbial inoculum and a parasite community treatment. The gut microbiome inocula were pre-exposed to the same parasite communities or a control treatment. Daphnia performance was monitored, and amplicon sequencing was performed to characterize the gut microbial community. Our experimental results showed that the gut microbiome plays no role in Daphnia tolerance upon parasite re-exposure. We did, however, find a main effect of the gut microbiome on Daphnia body size reflecting parasite specific responses. Our results also showed that it is rather the Daphnia genotype, and not the gut microbiome, that affected parasite-induced host mortality. Additionally, we found a role of the genotype in structuring the gut microbial community, both in alpha diversity as in the microbial composition.
APA, Harvard, Vancouver, ISO, and other styles
29

Castillo, Anakena M., Kristin Saltonstall, Carlos F. Arias, Karina A. Chavarria, Luis A. Ramírez-Camejo, Luis C. Mejía, and Luis F. De León. "The Microbiome of Neotropical Water Striders and Its Potential Role in Codiversification." Insects 11, no. 9 (August 31, 2020): 578. http://dx.doi.org/10.3390/insects11090578.

Full text
Abstract:
Insects host a highly diverse microbiome, which plays a crucial role in insect life. However, the composition and diversity of microbiomes associated with Neotropical freshwater insects is virtually unknown. In addition, the extent to which diversification of this microbiome is associated with host phylogenetic divergence remains to be determined. Here, we present the first comprehensive analysis of bacterial communities associated with six closely related species of Neotropical water striders in Panama. We used comparative phylogenetic analyses to assess associations between dominant bacterial linages and phylogenetic divergence among species of water striders. We found a total of 806 16S rRNA amplicon sequence variants (ASVs), with dominant bacterial taxa belonging to the phyla Proteobacteria (76.87%) and Tenericutes (19.51%). Members of the α- (e.g., Wolbachia) and γ- (e.g., Acinetobacter, Serratia) Proteobacteria, and Mollicutes (e.g., Spiroplasma) were predominantly shared across species, suggesting the presence of a core microbiome in water striders. However, some bacterial lineages (e.g., Fructobacillus, Fluviicola and Chryseobacterium) were uniquely associated with different water strider species, likely representing a distinctive feature of each species’ microbiome. These findings indicate that both host identity and environmental context are important drivers of microbiome diversity in water striders. In addition, they suggest that diversification of the microbiome is associated with diversification in water striders. Although more research is needed to establish the evolutionary consequences of host-microbiome interaction in water striders, our findings support recent work highlighting the role of bacterial community host-microbiome codiversification.
APA, Harvard, Vancouver, ISO, and other styles
30

Focà, Alfredo, Maria Carla Liberto, Angela Quirino, Nadia Marascio, Emilia Zicca, and Grazia Pavia. "Gut Inflammation and Immunity: What Is the Role of the Human Gut Virome?" Mediators of Inflammation 2015 (2015): 1–7. http://dx.doi.org/10.1155/2015/326032.

Full text
Abstract:
The human virome comprises viruses that infect host cells, virus-derived elements in our chromosomes, and viruses that infect other organisms, including bacteriophages and plant viruses. The development of high-throughput sequencing techniques has shown that the human gut microbiome is a complex community in which the virome plays a crucial role into regulation of intestinal immunity and homeostasis. Nevertheless, the size of the human virome is still poorly understood. Indeed the enteric virome is in a continuous and dynamic equilibrium with other components of the gut microbiome and the gut immune system, an interaction that may influence the health and disease of the host. We review recent evidence on the viruses found in the gastrointestinal tract, discussing their interactions with the resident bacterial microbiota and the host immune system, in order to explore the potential impact of the virome on human health.
APA, Harvard, Vancouver, ISO, and other styles
31

Reiman, Derek, Brian T. Layden, and Yang Dai. "MiMeNet: Exploring microbiome-metabolome relationships using neural networks." PLOS Computational Biology 17, no. 5 (May 17, 2021): e1009021. http://dx.doi.org/10.1371/journal.pcbi.1009021.

Full text
Abstract:
The advance in microbiome and metabolome studies has generated rich omics data revealing the involvement of the microbial community in host disease pathogenesis through interactions with their host at a metabolic level. However, the computational tools to uncover these relationships are just emerging. Here, we present MiMeNet, a neural network framework for modeling microbe-metabolite relationships. Using ten iterations of 10-fold cross-validation on three paired microbiome-metabolome datasets, we show that MiMeNet more accurately predicts metabolite abundances (mean Spearman correlation coefficients increase from 0.108 to 0.309, 0.276 to 0.457, and -0.272 to 0.264) and identifies more well-predicted metabolites (increase in the number of well-predicted metabolites from 198 to 366, 104 to 143, and 4 to 29) compared to state-of-art linear models for individual metabolite predictions. Additionally, we demonstrate that MiMeNet can group microbes and metabolites with similar interaction patterns and functions to illuminate the underlying structure of the microbe-metabolite interaction network, which could potentially shed light on uncharacterized metabolites through “Guilt by Association”. Our results demonstrated that MiMeNet is a powerful tool to provide insights into the causes of metabolic dysregulation in disease, facilitating future hypothesis generation at the interface of the microbiome and metabolomics.
APA, Harvard, Vancouver, ISO, and other styles
32

Zinöcker, Marit, and Inge Lindseth. "The Western Diet–Microbiome-Host Interaction and Its Role in Metabolic Disease." Nutrients 10, no. 3 (March 17, 2018): 365. http://dx.doi.org/10.3390/nu10030365.

Full text
Abstract:
The dietary pattern that characterizes the Western diet is strongly associated with obesity and related metabolic diseases, but biological mechanisms supporting these associations remain largely unknown. We argue that the Western diet promotes inflammation that arises from both structural and behavioral changes in the resident microbiome. The environment created in the gut by ultra-processed foods, a hallmark of the Western diet, is an evolutionarily unique selection ground for microbes that can promote diverse forms of inflammatory disease. Recognizing the importance of the microbiome in the development of diet-related disease has implications for future research, public dietary advice as well as food production practices. Research into food patterns suggests that whole foods are a common denominator of diets associated with a low level of diet-related disease. Hence, by studying how ultra-processing changes the properties of whole foods and how these foods affect the gut microbiome, more useful dietary guidelines can be made. Innovations in food production should be focusing on enabling health in the super-organism of man and microbe, and stronger regulation of potentially hazardous components of food products is warranted.
APA, Harvard, Vancouver, ISO, and other styles
33

Koyama, Motoko, and Geoffrey R. Hill. "The primacy of gastrointestinal tract antigen-presenting cells in lethal graft-versus-host disease." Blood 134, no. 24 (December 12, 2019): 2139–48. http://dx.doi.org/10.1182/blood.2019000823.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

Adolf, Lea A., and Simon Heilbronner. "Nutritional Interactions between Bacterial Species Colonising the Human Nasal Cavity: Current Knowledge and Future Prospects." Metabolites 12, no. 6 (May 27, 2022): 489. http://dx.doi.org/10.3390/metabo12060489.

Full text
Abstract:
The human nasal microbiome can be a reservoir for several pathogens, including Staphylococcus aureus. However, certain harmless nasal commensals can interfere with pathogen colonisation, an ability that could be exploited to prevent infection. Although attractive as a prophylactic strategy, manipulation of nasal microbiomes to prevent pathogen colonisation requires a better understanding of the molecular mechanisms of interaction that occur between nasal commensals as well as between commensals and pathogens. Our knowledge concerning the mechanisms of pathogen exclusion and how stable community structures are established is patchy and incomplete. Nutrients are scarce in nasal cavities, which makes competitive or mutualistic traits in nutrient acquisition very likely. In this review, we focus on nutritional interactions that have been shown to or might occur between nasal microbiome members. We summarise concepts of nutrient release from complex host molecules and host cells as well as of intracommunity exchange of energy-rich fermentation products and siderophores. Finally, we discuss the potential of genome-based metabolic models to predict complex nutritional interactions between members of the nasal microbiome.
APA, Harvard, Vancouver, ISO, and other styles
35

Kustrimovic, Natasa, Raffaella Bombelli, Denisa Baci, and Lorenzo Mortara. "Microbiome and Prostate Cancer: A Novel Target for Prevention and Treatment." International Journal of Molecular Sciences 24, no. 2 (January 12, 2023): 1511. http://dx.doi.org/10.3390/ijms24021511.

Full text
Abstract:
Growing evidence of the microbiome’s role in human health and disease has emerged since the creation of the Human Microbiome Project. Recent studies suggest that alterations in microbiota composition (dysbiosis) may play an essential role in the occurrence, development, and prognosis of prostate cancer (PCa), which remains the second most frequent male malignancy worldwide. Current advances in biological technologies, such as high-throughput sequencing, transcriptomics, and metabolomics, have enabled research on the gut, urinary, and intra-prostate microbiome signature and the correlation with local and systemic inflammation, host immunity response, and PCa progression. Several microbial species and their metabolites facilitate PCa insurgence through genotoxin-mediated mutagenesis or by driving tumor-promoting inflammation and dysfunctional immunosurveillance. However, the impact of the microbiome on PCa development, progression, and response to treatment is complex and needs to be fully understood. This review addresses the current knowledge on the host–microbe interaction and the risk of PCa, providing novel insights into the intraprostatic, gut, and urinary microbiome mechanisms leading to PCa carcinogenesis and treatment response. In this paper, we provide a detailed overview of diet changes, gut microbiome, and emerging therapeutic approaches related to the microbiome and PCa. Further investigation on the prostate-related microbiome and large-scale clinical trials testing the efficacy of microbiota modulation approaches may improve patient outcomes while fulfilling the literature gap of microbial–immune–cancer-cell mechanistic interactions.
APA, Harvard, Vancouver, ISO, and other styles
36

Ji, Dongxu, Hao Sun, Weichao Yang, Mingfu Gao, and Hui Xu. "Transfer of Human Microbiome to Drosophila Gut Model." Microorganisms 10, no. 3 (March 3, 2022): 553. http://dx.doi.org/10.3390/microorganisms10030553.

Full text
Abstract:
Laboratory animals with human microbiome have increasingly been used to study the role of bacteria and host interaction. Drosophila melanogaster, as a model of microbiota-host interaction with high reproductive efficiency and high availability, has always been lacking studies of interaction with human gut microbiome. In this study, we attempted to use antibiotic therapy and human fecal exposure strategy to transfer the human microbiome to the drosophila. The method includes depleting the original intestinal bacteria using a broad-spectrum antibiotic and then introducing human microorganisms by a diet supplemented with donor’s fecal samples. The sequencing results showed that 80–87.5% of the OTUs (Operational Taxonomic Units) from donor feces were adopted by the recipient drosophila following 30 days of observation. In comparison to females, the male recipient drosophila inherited more microbiota from the donor feces and had significantly increased lifespan as well as improved vertical climbing ability. Furthermore, distinctly differential expression patterns for age and insulin-like signaling-related genes were obtained for the male vs. female recipients. Only the male drosophila offspring acquired the characteristics of the donor fecal microbiota.
APA, Harvard, Vancouver, ISO, and other styles
37

Boesch, Maximilian, Lena Horvath, Florent Baty, Andreas Pircher, Dominik Wolf, Stephan Spahn, Ravid Straussman, Herbert Tilg, and Martin H. Brutsche. "Compartmentalization of the host microbiome: how tumor microbiota shapes checkpoint immunotherapy outcome and offers therapeutic prospects." Journal for ImmunoTherapy of Cancer 10, no. 11 (November 2022): e005401. http://dx.doi.org/10.1136/jitc-2022-005401.

Full text
Abstract:
The host microbiome is polymorphic, compartmentalized, and composed of distinctive tissue microbiomes. While research in the field of cancer immunotherapy has provided an improved understanding of the interaction with the gastrointestinal microbiome, the significance of the tumor-associated microbiome has only recently been grasped. This article provides a state-of-the-art review about the tumor-associated microbiome and sheds light on how local tumor microbiota shapes anticancer immunity and influences checkpoint immunotherapy outcome. The direct route of interaction between cancer cells, immune cells, and microbiota in the tumor microenvironment is emphasized and advocates a focus on the tumor-associated microbiome in addition to the spatially separated gut compartment. Since the mechanisms underlying checkpoint immunotherapy modulation by tumor-associated microbiota remain largely elusive, future research should dissect the pathways involved and outline strategies to therapeutically modulate microbes and their products within the tumor microenvironment. A more detailed knowledge about the mechanisms governing the composition and functional quality of the tumor microbiome will improve cancer immunotherapy and advance precision medicine for solid tumors.
APA, Harvard, Vancouver, ISO, and other styles
38

Scotti, Elena, Stéphanie Boué, Giuseppe Lo Sasso, Filippo Zanetti, Vincenzo Belcastro, Carine Poussin, Nicolas Sierro, et al. "Exploring the microbiome in health and disease." Toxicology Research and Application 1 (January 1, 2017): 239784731774188. http://dx.doi.org/10.1177/2397847317741884.

Full text
Abstract:
The analysis of human microbiome is an exciting and rapidly expanding field of research. In the past decade, the biological relevance of the microbiome for human health has become evident. Microbiome comprises a complex collection of microorganisms, with their genes and metabolites, colonizing different body niches. It is now well known that the microbiome interacts with its host, assisting in the bioconversion of nutrients and detoxification, supporting immunity, protecting against pathogenic microbes, and maintaining health. Remarkable new findings showed that our microbiome not only primarily affects the health and function of the gastrointestinal tract but also has a strong influence on general body health through its close interaction with the nervous system and the lung. Therefore, a perfect and sensitive balanced interaction of microbes with the host is required for a healthy body. In fact, growing evidence suggests that the dynamics and function of the indigenous microbiota can be influenced by many factors, including genetics, diet, age, and toxicological agents like cigarette smoke, environmental contaminants, and drugs. The disruption of this balance, that is called dysbiosis, is associated with a plethora of diseases, including metabolic diseases, inflammatory bowel disease, chronic obstructive pulmonary disease, periodontitis, skin diseases, and neurological disorders. The importance of the host microbiome for the human health has also led to the emergence of novel therapeutic approaches focused on the intentional manipulation of the microbiota, either by restoring missing functions or eliminating harmful roles. In the present review, we outline recent studies devoted to elucidate not only the role of microbiome in health conditions and the possible link with various types of diseases but also the influence of various toxicological factors on the microbial composition and function.
APA, Harvard, Vancouver, ISO, and other styles
39

Greenwood, Matthew P., Kelvin L. Hull, Marissa Brink-Hull, Melissa Lloyd, and Clint Rhode. "Feed and Host Genetics Drive Microbiome Diversity with Resultant Consequences for Production Traits in Mass-Reared Black Soldier Fly (Hermetia illucens) Larvae." Insects 12, no. 12 (December 1, 2021): 1082. http://dx.doi.org/10.3390/insects12121082.

Full text
Abstract:
Mass rearing the black soldier fly, Hermetia illucens, for waste bioremediation and valorisation is gaining traction on a global scale. While the health and productivity of this species are underpinned by associations with microbial taxa, little is known about the factors that govern gut microbiome assembly, function, and contributions towards host phenotypic development in actively feeding larvae. In the present study, a 16S rDNA gene sequencing approach applied to a study system incorporating both feed substrate and genetic variation is used to address this knowledge gap. It is determined that the alpha diversity of larval gut bacterial communities is driven primarily by features of the larval feed substrate, including the diversity of exogenous bacterial populations. Microbiome beta diversity, however, demonstrated patterns of differentiation consistent with an influence of diet, larval genetic background, and a potential interaction between these factors. Moreover, evidence for an association between microbiome structure and the rate of larval fat accumulation was uncovered. Taxonomic enrichment analysis and clustering of putative functional gut profiles further suggested that feed-dependent turnover in microbiome communities is most likely to impact larval characteristics. Taken together, these findings indicate that host–microbiome interactions in this species are complex yet relevant to larval trait emergence.
APA, Harvard, Vancouver, ISO, and other styles
40

Nurmi, Erika, Jude McElroy, Gerhard Hellemann, Christopher Laughlin, James McCracken, Chadi Calarge, and Lauren Seaman. "219. Host-Microbiome Interaction: A Putative Mechanism of Antipsychotic-Induced Weight Gain." Biological Psychiatry 85, no. 10 (May 2019): S91. http://dx.doi.org/10.1016/j.biopsych.2019.03.233.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

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

Full text
Abstract:
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
APA, Harvard, Vancouver, ISO, and other styles
42

Tisserant, Constance, and Arne Weiberg. "Extracellular vesicles in plant host-microbe interaction." How cells communicate - an introduction to extracellular vesicles 1, no. 1 (November 28, 2019): 46–50. http://dx.doi.org/10.47184/tev.2019.01.07.

Full text
Abstract:
Extracellular vesicles (EVs) are secreted lipid bilayer membrane particles that are increasingly drawing attention due to their potential role in intercellular communication. EVs have been mainly reported in mammalian systems, but are also found in non-mammalian classes, such as Archeae, bacteria, fungi, oomycetes, protozoa, invertebrates and plants. Over the last decade, EV research on mammalian systems has been massively advanced driven by the interests and applications of the biomedical field, while research on non-mammalian EVs that aims to understand the biological origins and functions of EVs remains rather descriptive and premature. Nevertheless, recent pioneering works resulted in novel concepts that place EVs carrying regulatory small RNAs as central players in inter-species and cross-kingdom communication with emphasis on host-pathogen, host-parasite and host-microbiome interactions. EVs transport small RNAs from microbe/pathogen/parasite to animal or plant hosts, and vice versa, which results in the manipulation of host immunity or microbial virulence, respectively. This article highlights some of the latest discoveries regarding EV-mediated communication across species and kingdoms with a special focus on plants and their interacting microbes.
APA, Harvard, Vancouver, ISO, and other styles
43

Beale, David J., Thao V. Nguyen, Rohan M. Shah, Andrew Bissett, Akhikun Nahar, Matthew Smith, Viviana Gonzalez-Astudillo, Christoph Braun, Brenda Baddiley, and Suzanne Vardy. "Host–Gut Microbiome Metabolic Interactions in PFAS-Impacted Freshwater Turtles (Emydura macquarii macquarii)." Metabolites 12, no. 8 (August 16, 2022): 747. http://dx.doi.org/10.3390/metabo12080747.

Full text
Abstract:
Per-and polyfluoroalkyl substances (PFAS) are a growing concern for humans, wildlife, and more broadly, ecosystem health. Previously, we characterised the microbial and biochemical impact of elevated PFAS on the gut microbiome of freshwater turtles (Emydura macquarii macquarii) within a contaminated catchment in Queensland, Australia. However, the understanding of PFAS impacts on this species and other aquatic organisms is still very limited, especially at the host–gut microbiome molecular interaction level. To this end, the present study aimed to apply these leading-edge omics technologies within an integrated framework that provides biological insight into the host turtle–turtle gut microbiome interactions of PFAS-impacted wild-caught freshwater turtles. For this purpose, faecal samples from PFAS-impacted turtles (n = 5) and suitable PFAS-free reference turtles (n = 5) were collected and analysed. Data from 16S rRNA gene amplicon sequencing and metabolomic profiling of the turtle faeces were integrated using MetOrigin to assign host, microbiome, and co-metabolism activities. Significant variation in microbial composition was observed between the two turtle groups. The PFAS-impacted turtles showed a higher relative abundance of Firmicutes and a lower relative abundance of Bacteroidota than the reference turtles. The faecal metabolome showed several metabolites and pathways significantly affected by PFAS exposure. Turtles exposed to PFAS displayed altered amino acid and butanoate metabolisms, as well as altered purine and pyrimidine metabolism. It is predicted from this study that PFAS-impacted both the metabolism of the host turtle and its gut microbiota which in turn has the potential to influence the host’s physiology and health.
APA, Harvard, Vancouver, ISO, and other styles
44

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

Full text
Abstract:
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.
APA, Harvard, Vancouver, ISO, and other styles
45

Verma, Sargam, Lucas Carvalho Basilio Azevedo, Jyoti Pandey, Saksham Khusharia, Madhuree Kumari, Dharmendra Kumar, Kaushalendra, Nikunj Bhardwaj, Pratibha Teotia, and Ajay Kumar. "Microbial Intervention: An Approach to Combat the Postharvest Pathogens of Fruits." Plants 11, no. 24 (December 9, 2022): 3452. http://dx.doi.org/10.3390/plants11243452.

Full text
Abstract:
Plants host diverse microbial communities, which undergo a complex interaction with each other. Plant-associated microbial communities provide various benefits to the host directly or indirectly, viz. nutrient acquisition, protection from pathogen invaders, mitigation from different biotic and abiotic stress. Presently, plant-associated microbial strains are frequently utilized as biofertilizers, biostimulants and biocontrol agents in greenhouse and field conditions and have shown satisfactory results. Nowadays, the plant/fruit microbiome has been employed to control postharvest pathogens and postharvest decay, and to maintain the quality or shelf life of fruits. In this context, the intervention of the natural fruit microbiome or the creation of synthetic microbial communities to modulate the functional attributes of the natural microbiome is an emerging aspect. In this regard, we discuss the community behavior of microbes in natural conditions and how the microbiome intervention plays a crucial role in the postharvest management of fruits.
APA, Harvard, Vancouver, ISO, and other styles
46

Cho, Young-Dan, Kyoung-Hwa Kim, Yong-Moo Lee, Young Ku, and Yang-Jo Seol. "Oral Microbiome and Host Health: Review on Current Advances in Genome-Wide Analysis." Applied Sciences 11, no. 9 (April 29, 2021): 4050. http://dx.doi.org/10.3390/app11094050.

Full text
Abstract:
The oral microbiome is an important part of the human microbiome. The oral cavity has the second largest microbiota after the intestines, and its open structure creates a special environment. With the development of technology such as next-generation sequencing and bioinformatics, extensive in-depth microbiome studies have become possible. They can also be applied in the clinical field in terms of diagnosis and treatment. Many microbiome studies have been performed on oral and systemic diseases, showing a close association between the two. Understanding the oral microbiome and host interaction is expected to provide future directions to explore the functional and metabolic changes in diseases, and to uncover the molecular mechanisms for drug development and treatment that facilitate personalized medicine. The aim of this review was to provide comprehension regarding research trends in oral microbiome studies and establish the link between oral microbiomes and systemic diseases based on the latest technique of genome-wide analysis.
APA, Harvard, Vancouver, ISO, and other styles
47

Larsen, Peter E., and Yang Dai. "Modeling interaction networks between host, diet, and bacteria predicts obesogenesis in a mouse model." Frontiers in Molecular Biosciences 9 (November 15, 2022). http://dx.doi.org/10.3389/fmolb.2022.1059094.

Full text
Abstract:
Host-microbiome interactions are known to have substantial effects on human health, but the diversity of the human microbiome makes it difficult to definitively attribute specific microbiome features to a host phenotype. One approach to overcoming this challenge is to use animal models of host-microbiome interaction, but it must be determined that relevant aspects of host-microbiome interactions are reflected in the animal model. One such experimental validation is an experiment by Ridura et al. In that experiment, transplanting a microbiome from a human into a mouse also conferred the human donor’s obesity phenotype. We have aggregated a collection of previously published host-microbiome mouse-model experiments and combined it with thousands of sequenced and annotated bacterial genomes and metametabolomic pathways. Three computational models were generated, each model reflecting an aspect of host-microbiome interactions: 1) Predict the change in microbiome community structure in response to host diet using a community interaction network, 2) Predict metagenomic data from microbiome community structure, and 3) Predict host obesogenesis from modeled microbiome metagenomic data. These computationally validated models were combined into an integrated model of host-microbiome-diet interactions and used to replicate the Ridura experiment in silico. The results of the computational models indicate that network-based models are significantly more predictive than similar but non-network-based models. Network-based models also provide additional insight into the molecular mechanisms of host-microbiome interaction by highlighting metabolites and metabolic pathways proposed to be associated with microbiome-based obesogenesis. While the models generated in this study are likely too specific to the animal models and experimental conditions used to train our models to be of general utility in a broader understanding of obesogenesis, the approach detailed here is expected to be a powerful tool of investigating multiple types of host-microbiome interactions.
APA, Harvard, Vancouver, ISO, and other styles
48

Cox, Timothy O., Patrick Lundgren, Kirti Nath, and Christoph A. Thaiss. "Metabolic control by the microbiome." Genome Medicine 14, no. 1 (July 29, 2022). http://dx.doi.org/10.1186/s13073-022-01092-0.

Full text
Abstract:
AbstractThe interaction between the metabolic activities of the intestinal microbiome and its host forms an important part of health. The basis of this interaction is in part mediated by the release of microbially-derived metabolites that enter the circulation. These products of microbial metabolism thereby interface with the immune, metabolic, or nervous systems of the host to influence physiology. Here, we review the interactions between the metabolic activities of the microbiome and the systemic metabolism of the host. The concept that the endocrine system includes more than just the eukaryotic host component enables the rational design of exogenous interventions that shape human metabolism. An improved mechanistic understanding of the metabolic microbiome-host interaction may therefore pioneer actionable microbiota-based diagnostics or therapeutics that allow the control of host systemic metabolism via the microbiome.
APA, Harvard, Vancouver, ISO, and other styles
49

Dong, Qiang, Eric S. Chen, Chen Zhao, and Chengcheng Jin. "Host-Microbiome Interaction in Lung Cancer." Frontiers in Immunology 12 (May 24, 2021). http://dx.doi.org/10.3389/fimmu.2021.679829.

Full text
Abstract:
Commensal microbiota has emerged as an essential biomarker and regulator of both tumorigenesis and response to cancer therapy. However, our current knowledge about microbiota in cancer has been largely limited to intestinal microbiota. As a mucosal organ harboring one of the largest surface areas in the body, the lung is exposed to a variety of microbes through inhalation and micro-aspiration, and is colonized by a diverse bacterial community in both physiological and pathological conditions. Importantly, increasing evidence has linked the lung microbiome to cancer development. Studies in lung cancer patients and mouse models have revealed tumor-associated dysregulation of the local microbiome in the lung, which in turn impacts cancer progression by shaping the tumor microenvironment and modulating the activity of tumor-infiltrating immune cells. These findings not only provide novel mechanistic insight into the biology of lung cancer but also shed light on new therapeutic targets and strategies for lung cancer prevention and treatment. The goal of this review is to discuss the key findings, remaining questions, and future directions in this new and exciting field.
APA, Harvard, Vancouver, ISO, and other styles
50

Cangioli, Lisa, Alice Checcucci, Alessio Mengoni, and Camilla Fagorzi. "Legume tasters: symbiotic rhizobia host preference and smart inoculant formulations." Biological Communications 66, no. 1 (March 31, 2021). http://dx.doi.org/10.21638/spbu03.2021.106.

Full text
Abstract:
Mutualistic interactions have great importance in ecology, with genetic information that takes shape through interactions within the symbiotic partners and between the partners and the environment. It is known that variation of the host-associated microbiome contributes to buffer adaptation challenges of the host’s physiology when facing varying environmental conditions. In agriculture, pivotal examples are symbiotic nitrogen-fixing rhizobia, known to contribute greatly to host (legume plants) adaptation and host productivity. A holistic view of increasing crop yield and resistance to biotic and abiotic stresses is that of microbiome engineering, the exploitation of a host-associated microbiome through its rationally designed manipulation with synthetic microbial communities. However, several studies highlighted that the expression of the desired phenotype in the host resides in species-specific, even genotype-specific interactions between the symbiotic partners. Consequently, there is a need to dissect such an intimate level of interaction, aiming to identify the main genetic components in both partners playing a role in symbiotic differences/host preferences. In the present paper, while briefly reviewing the knowledge and the challenges in plant–microbe interaction and rhizobial studies, we aim to promote research on genotype x genotype interaction between rhizobia and host plants for a rational design of synthetic symbiotic nitrogen-fixing microbial communities to be used for sustainably improving leguminous plants yield.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography