Статті в журналах з теми "Host - microbial interaction"

Щоб переглянути інші типи публікацій з цієї теми, перейдіть за посиланням: Host - microbial interaction.

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся з топ-50 статей у журналах для дослідження на тему "Host - microbial interaction".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Переглядайте статті в журналах для різних дисциплін та оформлюйте правильно вашу бібліографію.

1

Priya, Ayushi. "Microbial Host Interaction in Periodontal Diseases." Indian Journal of Public Health Research & Development 10, no. 11 (2019): 798. http://dx.doi.org/10.5958/0976-5506.2019.03583.6.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Fasano, Alessio. "Understanding the Dialogue: the Microbial–Host Interaction." Annales Nestlé (English ed.) 67, no. 1 (2009): 9–18. http://dx.doi.org/10.1159/000187165.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

BUZA, Victoria, Maria Catalina MATEI, and Laura Cristina STEFANUT. "Intestinal Ecosystem: Interaction and Coexistence Between “Parasitome” and Microbial Communities." Bulletin of University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca. Veterinary Medicine 77, no. 1 (June 3, 2020): 15. http://dx.doi.org/10.15835/buasvmcn-vm:2019.0032.

Повний текст джерела
Анотація:
The vertebrate gut has been continuously populated with complex and dynamic microbial and eukaryotic communities, that over millions of years have coevolved both spatially and temporally (Kreisinger et al., 2015). Due to the prolonged coexistence, intestinal parasites (protozoa and helminths) and resident microbiota have developed the ability to influence one another by several mechanisms: 1) produce changes at the level of intestinal mucus and epithelial barrier, 2) alter the host immune response or 3) direct interaction (Leung et al., 2018). The uncontrolled use of anthelmintics can lead to the elimination of commensal organisms and alteration of host immunity and intestinal microbial community composition. Thus, the aim of this research is to highlight the complexity of interactions between intestinal bacteria and parasites and their importance for the host. The “parasitome”- microbiota relationship is a complex phenomenon that plays an essential role in host intestinal homeostasis, the absence or alteration of either of these organisms being able to cause a severe disruption of host immune system (Leung et al., 2018). Is therefore essential to acquire a deeper understanding of the molecular mechanisms of interaction between these two communities.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Zhang, Rui, and Aixin Hou. "Host-Microbe Interactions in Caenorhabditis elegans." ISRN Microbiology 2013 (August 1, 2013): 1–7. http://dx.doi.org/10.1155/2013/356451.

Повний текст джерела
Анотація:
A good understanding of how microbes interact with hosts has a direct bearing on our capability of fighting infectious microbial pathogens and making good use of beneficial ones. Among the model organisms used to study reciprocal actions among microbes and hosts, C. elegans may be the most advantageous in the context of its unique attributes such as the short life cycle, easiness of laboratory maintenance, and the availability of different genetic mutants. This review summarizes the recent advances in understanding host-microbe interactions in C. elegans. Although these investigations have greatly enhanced our understanding of C. elegans-microbe relationships, all but one of them involve only one or few microbial species. We argue here that more research is needed for exploring the evolution and establishment of a complex microbial community in the worm’s intestine and its interaction with the host.
Стилі APA, Harvard, Vancouver, ISO та ін.
5

NV, Beloborodova. "Low-Molecular Weight Bacterial Metabolites in Host-Microbial Interaction." Infectious & Non Infectious Diseases 2, no. 1 (June 9, 2016): 1–11. http://dx.doi.org/10.24966/inid-8654/100011.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Kumar, P. S., M. F. Monteiro, S. M. Dabdoub, G. L. Miranda, M. Z. Casati, F. V. Ribeiro, F. R. Cirano, S. P. Pimentel, and R. C. V. Casarin. "Subgingival Host-Microbial Interactions in Hyperglycemic Individuals." Journal of Dental Research 99, no. 6 (March 16, 2020): 650–57. http://dx.doi.org/10.1177/0022034520906842.

Повний текст джерела
Анотація:
Type 2 diabetes mellitus (T2DM) is an established risk factor for periodontitis, yet its contribution to creating host-bacterial disequilibrium in the subgingival crevice is poorly understood. The present investigation aimed to quantify the impact of hyperglycemia on host-bacterial interactions in established periodontitis and to map shifts in these dynamics following mechanical nonsurgical therapy. Seventeen T2DM and 17 non-T2DM subjects with generalized severe chronic periodontitis were recruited along with 20 periodontally healthy individuals. Subjects with periodontitis were treated with scaling and root planing (SRP). Samples of subgingival biofilm and gingival crevicular fluid were collected at baseline and at 1-, 3-, and 6 mo postoperatively. Correlations were generated between 13.7 million 16S ribosomal DNA sequences and 8 immune mediators. Intermicrobial and host-microbial interactions were modeled using differential network analysis. Periodontal health was characterized by a sparse interbacterial and highly connected cytokine-bacterial network, while both normoglycemics and T2DM subjects with periodontitis demonstrated robust congeneric and intergeneric hubs but significantly fewer cytokine-bacterial connections. Following SRP, the cytokine-bacterial edges demonstrated a 2-fold increase 1 mo postoperatively and a 10-fold increase at 6 mo in normoglycemics. In hyperglycemics, there was a doubling at 1 mo but no further changes thereafter. These shifts accompanied an increasingly sparse interbacterial network. In normoglycemics, the nodes anchored by interleukin (IL)–4, IL-6, and IL-10 demonstrated greatest rewiring, while in hyperglycemics, IL-1β, IL-6, INF-γ, and IL-17 exhibited progressive rewiring. Thus, the present investigation points to a breakdown in host-bacterial mutualism in periodontitis, with interbacterial interactions rather than host-bacterial interactions primarily determining community assembly. Hyperglycemia further exacerbates this uncoupled mutualism. Our data also demonstrate that while nonsurgical therapy might not consistently alter microbial abundances or lower proinflammatory molecules, it “reboots” the interaction between the immunoinflammatory system and the newly colonizing microbiome, restoring a role for the immune system in determining bacterial colonization. However, this outcome is lower and delayed in hyperglycemics.
Стилі APA, Harvard, Vancouver, ISO та ін.
7

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.

Повний текст джерела
Анотація:
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.
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Weiland-Bräuer, Nancy. "Friends or Foes—Microbial Interactions in Nature." Biology 10, no. 6 (June 2, 2021): 496. http://dx.doi.org/10.3390/biology10060496.

Повний текст джерела
Анотація:
Microorganisms are present in nearly every niche on Earth and mainly do not exist solely but form communities of single or mixed species. Within such microbial populations and between the microbes and a eukaryotic host, various microbial interactions take place in an ever-changing environment. Those microbial interactions are crucial for a successful establishment and maintenance of a microbial population. The basic unit of interaction is the gene expression of each organism in this community in response to biotic or abiotic stimuli. Differential gene expression is responsible for producing exchangeable molecules involved in the interactions, ultimately leading to community behavior. Cooperative and competitive interactions within bacterial communities and between the associated bacteria and the host are the focus of this review, emphasizing microbial cell–cell communication (quorum sensing). Further, metagenomics is discussed as a helpful tool to analyze the complex genomic information of microbial communities and the functional role of different microbes within a community and to identify novel biomolecules for biotechnological applications.
Стилі APA, Harvard, Vancouver, ISO та ін.
9

Galiana, Eric, Antoine Marais, Catherine Mura, Benoît Industri, Gilles Arbiol, and Michel Ponchet. "Ecosystem Screening Approach for Pathogen-Associated Microorganisms Affecting Host Disease." Applied and Environmental Microbiology 77, no. 17 (July 8, 2011): 6069–75. http://dx.doi.org/10.1128/aem.05371-11.

Повний текст джерела
Анотація:
ABSTRACTThe microbial community in which a pathogen evolves is fundamental to disease outcome. Species interacting with a pathogen on the host surface shape the distribution, density, and genetic diversity of the inoculum, but the role of these species is rarely determined. The screening method developed here can be used to characterize pathogen-associated species affecting disease. This strategy involves three steps: (i) constitution of the microbial community, using the pathogen as a trap; (ii) community selection, using extracts from the pathogen as the sole nutrient source; and (iii) molecular identification and the screening of isolates focusing on their effects on the growth of the pathogenin vitroand host disease. This approach was applied to a soilborne plant pathogen,Phytophthora parasitica, structured in a biofilm, for screening the microbial community from the rhizosphere ofNicotiana tabacum(the host). Two of the characterized eukaryotes interfered with the oomycete cycle and may affect the host disease. AVorticellaspecies acted through a mutualistic interaction withP. parasitica, disseminating pathogenic material by leaving the biofilm. APhomaspecies established an amensal interaction withP. parasitica, strongly suppressing disease by inhibitingP. parasiticagermination. This screening method is appropriate for all nonobligate pathogens. It allows the definition of microbial species as promoters or suppressors of a disease for a given biotope. It should also help to identify important microbial relationships for ecology and evolution of pathogens.
Стилі APA, Harvard, Vancouver, ISO та ін.
10

Casadevall, Arturo, and Liise-anne Pirofski. "Microbial virulence results from the interaction between host and microorganism." Trends in Microbiology 11, no. 4 (April 2003): 157–58. http://dx.doi.org/10.1016/s0966-842x(03)00008-8.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
11

Suzuki, Kenta, Masato S. Abe, Daiki Kumakura, Shinji Nakaoka, Fuki Fujiwara, Hirokuni Miyamoto, Teruno Nakaguma, et al. "Chemical-Mediated Microbial Interactions Can Reduce the Effectiveness of Time-Series-Based Inference of Ecological Interaction Networks." International Journal of Environmental Research and Public Health 19, no. 3 (January 22, 2022): 1228. http://dx.doi.org/10.3390/ijerph19031228.

Повний текст джерела
Анотація:
Network-based assessments are important for disentangling complex microbial and microbial–host interactions and can provide the basis for microbial engineering. There is a growing recognition that chemical-mediated interactions are important for the coexistence of microbial species. However, so far, the methods used to infer microbial interactions have been validated with models assuming direct species-species interactions, such as generalized Lotka–Volterra models. Therefore, it is unclear how effective existing approaches are in detecting chemical-mediated interactions. In this paper, we used time series of simulated microbial dynamics to benchmark five major/state-of-the-art methods. We found that only two methods (CCM and LIMITS) were capable of detecting interactions. While LIMITS performed better than CCM, it was less robust to the presence of chemical-mediated interactions, and the presence of trophic competition was essential for the interactions to be detectable. We show that the existence of chemical-mediated interactions among microbial species poses a new challenge to overcome for the development of a network-based understanding of microbiomes and their interactions with hosts and the environment.
Стилі APA, Harvard, Vancouver, ISO та ін.
12

He, X., F. Li, B. Bor, K. Koyano, L. Cen, X. Xiao, W. Shi, and D. T. W. Wong. "Human tRNA-Derived Small RNAs Modulate Host–Oral Microbial Interactions." Journal of Dental Research 97, no. 11 (April 27, 2018): 1236–43. http://dx.doi.org/10.1177/0022034518770605.

Повний текст джерела
Анотація:
Coevolution of the human host and its associated microbiota has led to sophisticated interactions to maintain a delicate homeostasis. Emerging evidence suggests that in addition to small molecules, peptides, and proteins, small regulatory noncoding RNAs (sRNAs) might play an important role in cross-domain interactions. In this study, we revealed the presence of diverse host transfer RNA–derived small RNAs (tsRNAs) among human salivary sRNAs. We selected 2 tsRNAs (tsRNA-000794 and tsRNA-020498) for further study based on their high sequence similarity to specific tRNAs from a group of Gram-negative oral bacteria, including Fusobacterium nucleatum, a key oral commensal and opportunistic pathogen. We showed that the presence of F. nucleatum triggers exosome-mediated release of tsRNA-000794 and tsRNA-020498 by human normal oral keratinocyte cells. Furthermore, both tsRNA candidates exerted a growth inhibition effect on F. nucleatum, likely through interference with bacterial protein biosynthesis, but did not affect the growth of Streptococcus mitis, a health-associated oral Gram-positive bacterium whose genome does not carry sequences bearing high similarity to either tsRNA. Our data provide the first line of evidence for the modulatory role of host-derived tsRNAs in the microbial-host interaction.
Стилі APA, Harvard, Vancouver, ISO та ін.
13

Yin, Jiayi, Fengcheng Li, Ying Zhou, Minjie Mou, Yinjing Lu, Kangli Chen, Jia Xue, et al. "INTEDE: interactome of drug-metabolizing enzymes." Nucleic Acids Research 49, no. D1 (October 12, 2020): D1233—D1243. http://dx.doi.org/10.1093/nar/gkaa755.

Повний текст джерела
Анотація:
Abstract Drug-metabolizing enzymes (DMEs) are critical determinant of drug safety and efficacy, and the interactome of DMEs has attracted extensive attention. There are 3 major interaction types in an interactome: microbiome–DME interaction (MICBIO), xenobiotics–DME interaction (XEOTIC) and host protein–DME interaction (HOSPPI). The interaction data of each type are essential for drug metabolism, and the collective consideration of multiple types has implication for the future practice of precision medicine. However, no database was designed to systematically provide the data of all types of DME interactions. Here, a database of the Interactome of Drug-Metabolizing Enzymes (INTEDE) was therefore constructed to offer these interaction data. First, 1047 unique DMEs (448 host and 599 microbial) were confirmed, for the first time, using their metabolizing drugs. Second, for these newly confirmed DMEs, all types of their interactions (3359 MICBIOs between 225 microbial species and 185 DMEs; 47 778 XEOTICs between 4150 xenobiotics and 501 DMEs; 7849 HOSPPIs between 565 human proteins and 566 DMEs) were comprehensively collected and then provided, which enabled the crosstalk analysis among multiple types. Because of the huge amount of accumulated data, the INTEDE made it possible to generalize key features for revealing disease etiology and optimizing clinical treatment. INTEDE is freely accessible at: https://idrblab.org/intede/
Стилі APA, Harvard, Vancouver, ISO та ін.
14

Feng, Xin, Caiyu Luo, and Jianwei Che. "Diet modulates host health through gut microbiota derived extracellular vesicles: A short review." Aceh Journal of Animal Science 8, no. 2 (May 25, 2023): 58–61. http://dx.doi.org/10.13170/ajas.8.2.32030.

Повний текст джерела
Анотація:
Gut microbes are involved with many host physiological processes including digestion, metabolism, immune response, gut function and behavior. Among all the factors, diet is being considered the most important one to modulate gut microbiota composition, metabolism and their metabolites. Extracellular vesicles (EVs) are secreted to the intestinal environment by gut microbes and play an essential role in gut microbe-host communication. This paper aims to review how diet affects gut microbial EVs and its composition as well as how this change further affects host health. This review summarizes the latest research progress of interaction among diet, gut microbial EVs, and host health. Through the microbiota-gut axis, gut microbial EVs involve in many physiological activities, including brain function, metabolism, gut function and immune response. It has been verified that diet composition has direct changes on gut microbial morphology and internal molecules within gut microbial EVs. Overall, studies investigating the effects of diet through gut microbial EVs on host health are very limited. Future research regarding axis of diet-gut microbial EVs-host health is recommended.
Стилі APA, Harvard, Vancouver, ISO та ін.
15

Hold, Georgina L., Indrani Mukhopadhya, and Tom P. Monie. "Innate Immune Sensors and Gastrointestinal Bacterial Infections." Clinical and Developmental Immunology 2011 (2011): 1–11. http://dx.doi.org/10.1155/2011/579650.

Повний текст джерела
Анотація:
The gastrointestinal microbiota is a major source of immune stimulation. The interaction between host pattern-recognition receptors and conserved microbial ligands profoundly influences infection dynamics. Identifying and understanding the nature of these interactions is a key step towards obtaining a clearer picture of microbial pathogenesis. These interactions underpin a complex interplay between microbe and host that has far reaching consequences for both. Here, we review the role of pattern recognition receptors in three prototype diseases affecting the stomach, the small intestine, and large intestine, respectively (Helicobacter pyloriinfection,Salmonellainfection, and inflammatory bowel disease). Specifically, we review the nature and impact of pathogen:receptor interactions, their impact upon pathogenesis, and address the relevance of pattern recognition receptors in the development of therapies for gastrointestinal diseases.
Стилі APA, Harvard, Vancouver, ISO та ін.
16

Snyder, Greg, Daniel Deredge, Anna Waldhuber, Theresa Fresquez, Patrick Smith, Suri Duerr, Christine Cirl, et al. "Development of microbial-derived inhibitory peptides using structural studies of microbial TIR proteins TcpB, TcpC and host adapters TIRAP and MyD88. (INM9P.449)." Journal of Immunology 192, no. 1_Supplement (May 1, 2014): 189.2. http://dx.doi.org/10.4049/jimmunol.192.supp.189.2.

Повний текст джерела
Анотація:
Abstract Microbial pathogens have evolved mechanisms to regulate and evade innate immunity. One such mechanism involves the subversion of Toll-like receptor (TLR) signaling by bacterial TIR Interacting Proteins (TIPs). TIPs are thought to function by disruption of host Toll/IL-1 receptor (TIR) signaling proteins. For example, the TIP proteins TcpB from Brucella and TcpC from E. coli inhibit TLR signaling through direct interaction with host adapter proteins TIRAP and MyD88. We have previously reported the crystal structure of MyD88 and characterized TcpC peptides capable to inhibit TLR4 and MyD88 signaling. We now report the X-ray crystal structure of the Brucella TIR protein TcpB and characterize its interactions with TIRAP using hydrogen/ deuterium (H/D) exchange mass spectrometry, co-immunoprecipitation and NF-ΚB reporter assays. The crystal structure of TcpB reveals the BB loop microtubule-binding site as well as a symmetrical dimer involving the DD and EE loops. The dimer interface is further characterized through H/D exchange mass spectrometry, which identifies a set of candidate potential TcpB blocking peptides. A comparison between the microbial TcpB, TIRAP and MyD88 crystal structures reveal differences in the region that encompasses the BB loop. These findings lend insight into the molecular mechanisms of microbial mimicry of host signaling adapter proteins and provide a framework for identification and development of novel microbial-derived therapeutics.
Стилі APA, Harvard, Vancouver, ISO та ін.
17

You, Chuan, Dan Qin, Yumeng Wang, Wenyi Lan, Yehong Li, Baohong Yu, Yajun Peng, Jieru Xu, and Jinyan Dong. "Plant Triterpenoids Regulate Endophyte Community to Promote Medicinal Plant Schisandra sphenanthera Growth and Metabolites Accumulation." Journal of Fungi 7, no. 10 (September 23, 2021): 788. http://dx.doi.org/10.3390/jof7100788.

Повний текст джерела
Анотація:
Beneficial interactions between endophytes and plants are critical for plant growth and metabolite accumulation. Nevertheless, the secondary metabolites controlling the feedback between the host plant and the endophytic microbial community remain elusive in medicinal plants. In this report, we demonstrate that plant-derived triterpenoids predominantly promote the growth of endophytic bacteria and fungi, which in turn promote host plant growth and secondary metabolite productions. From culturable bacterial and fungal microbial strains isolated from the medicinal plant Schisandra sphenanthera, through triterpenoid-mediated screens, we constructed six synthetic communities (SynComs). By using a binary interaction method in plates, we revealed that triterpenoid-promoted bacterial and fungal strains (TPB and TPF) played more positive roles in the microbial community. The functional screening of representative strains suggested that TPB and TPF provide more beneficial abilities to the host. Moreover, pot experiments in a sterilized system further demonstrated that TPB and TPF play important roles in host growth and metabolite accumulation. In summary, these experiments revealed a role of triterpenoids in endophytic microbiome assembly and indicated a strategy for constructing SynComs on the basis of the screening of secondary metabolites, in which bacteria and fungi join forces to promote plant health. These findings may open new avenues towards the breeding of high yielding and high metabolite-accumulating medicinal plants by exploiting their interaction with beneficial endophytes.
Стилі APA, Harvard, Vancouver, ISO та ін.
18

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.

Повний текст джерела
Анотація:
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.
Стилі APA, Harvard, Vancouver, ISO та ін.
19

Jha, Yachana, Budheswar Dehury, S. P. Jeevan Kumar, Anurag Chaurasia, Udai B. Singh, Manoj Kumar Yadav, U. B. Angadi та ін. "Delineation of molecular interactions of plant growth promoting bacteria induced β-1,3-glucanases and guanosine triphosphate ligand for antifungal response in rice: a molecular dynamics approach". Molecular Biology Reports 49, № 4 (16 грудня 2021): 2579–89. http://dx.doi.org/10.1007/s11033-021-07059-5.

Повний текст джерела
Анотація:
Abstract Background The plant growth is influenced by multiple interactions with biotic (microbial) and abiotic components in their surroundings. These microbial interactions have both positive and negative effects on plant. Plant growth promoting bacterial (PGPR) interaction could result in positive growth under normal as well as in stress conditions. Methods Here, we have screened two PGPR’s and determined their potential in induction of specific gene in host plant to overcome the adverse effect of biotic stress caused by Magnaporthe grisea, a fungal pathogen that cause blast in rice. We demonstrated the glucanase protein mode of action by performing comparative modeling and molecular docking of guanosine triphosphate (GTP) ligand with the protein. Besides, molecular dynamic simulations have been performed to understand the behavior of the glucanase-GTP complex. Results The results clearly showed that selected PGPR was better able to induce modification in host plant at morphological, biochemical, physiological and molecular level by activating the expression of β-1,3-glucanases gene in infected host plant. The docking results indicated that Tyr75, Arg256, Gly258, and Ser223 of glucanase formed four crucial hydrogen bonds with the GTP, while, only Val220 found to form hydrophobic contact with ligand. Conclusions The PGPR able to induce β-1,3-glucanases gene in host plant upon pathogenic interaction and β-1,3-glucanases form complex with GTP by hydrophilic interaction for induction of defense cascade for acquiring resistance against Magnaporthe grisea. Graphical abstract
Стилі APA, Harvard, Vancouver, ISO та ін.
20

Todd, Olivia A., and Brian M. Peters. "Candida albicans and Staphylococcus aureus Pathogenicity and Polymicrobial Interactions: Lessons beyond Koch’s Postulates." Journal of Fungi 5, no. 3 (September 4, 2019): 81. http://dx.doi.org/10.3390/jof5030081.

Повний текст джерела
Анотація:
While Koch’s Postulates have established rules for microbial pathogenesis that have been extremely beneficial for monomicrobial infections, new studies regarding polymicrobial pathogenesis defy these standards. The explosion of phylogenetic sequence data has revolutionized concepts of microbial interactions on and within the host. However, there remains a paucity of functional follow-up studies to delineate mechanisms driven by such interactions and how they shape health or disease. That said, one particular microbial pairing, the fungal opportunist Candida albicans and the bacterial pathogen Staphylococcus aureus, has received much attention over the last decade. Therefore, the objective of this review is to discuss the multi-faceted mechanisms employed by these two ubiquitous human pathogens during polymicrobial growth, including how they: establish and persist in inter-Kingdom biofilms, tolerate antimicrobial therapy, co-invade host tissue, exacerbate quorum sensing and staphylococcal toxin production, and elicit infectious synergism. Commentary regarding new challenges and remaining questions related to future discovery of this fascinating fungal–bacterial interaction is also provided.
Стилі APA, Harvard, Vancouver, ISO та ін.
21

Lee, Sungeun, Ella T. Sieradzki, Alexa M. Nicolas, Robin L. Walker, Mary K. Firestone, Christina Hazard, and Graeme W. Nicol. "Methane-derived carbon flows into host–virus networks at different trophic levels in soil." Proceedings of the National Academy of Sciences 118, no. 32 (August 4, 2021): e2105124118. http://dx.doi.org/10.1073/pnas.2105124118.

Повний текст джерела
Анотація:
The concentration of atmospheric methane (CH4) continues to increase with microbial communities controlling soil–atmosphere fluxes. While there is substantial knowledge of the diversity and function of prokaryotes regulating CH4 production and consumption, their active interactions with viruses in soil have not been identified. Metagenomic sequencing of soil microbial communities enables identification of linkages between viruses and hosts. However, this does not determine if these represent current or historical interactions nor whether a virus or host are active. In this study, we identified active interactions between individual host and virus populations in situ by following the transfer of assimilated carbon. Using DNA stable-isotope probing combined with metagenomic analyses, we characterized CH4-fueled microbial networks in acidic and neutral pH soils, specifically primary and secondary utilizers, together with the recent transfer of CH4-derived carbon to viruses. A total of 63% of viral contigs from replicated soil incubations contained homologs of genes present in known methylotrophic bacteria. Genomic sequences of 13C-enriched viruses were represented in over one-third of spacers in CRISPR arrays of multiple closely related Methylocystis populations and revealed differences in their history of viral interaction. Viruses infecting nonmethanotrophic methylotrophs and heterotrophic predatory bacteria were also identified through the analysis of shared homologous genes, demonstrating that carbon is transferred to a diverse range of viruses associated with CH4-fueled microbial food networks.
Стилі APA, Harvard, Vancouver, ISO та ін.
22

Wu, Yuqi, Yufei Zheng, Yanan Chen, Gongwen Chen, Huoqing Zheng, and Fuliang Hu. "Apis cerana gut microbiota contribute to host health though stimulating host immune system and strengthening host resistance to Nosema ceranae." Royal Society Open Science 7, no. 5 (May 2020): 192100. http://dx.doi.org/10.1098/rsos.192100.

Повний текст джерела
Анотація:
Gut microbial communities play vital roles in the modulation of many insects' immunity, including Apis mellifera . However, little is known about the interaction of Apis cerana gut bacteria and A. cerana immune system. Here in this study, we conducted a comparison between germ-free gut microbiota deficient (GD) workers and conventional gut community (CV) workers, to reveal the possible impact of gut microbiota on the expression of A. cerana antimicrobial peptides and immune regulate pathways. We also test whether A. cerana gut microbiota can strengthen host resistance to Nosema ceranae . We find that the expression of apidaecin , abaecin and hymenoptaecin were significantly upregulated with the presence of gut bacteria, and JNK pathway was activated; in the meanwhile, the existence of gut bacteria inhibited the proliferation of Nosema ceranae . These demonstrated the essential role of A. cerana gut microbiota to host health and provided critical insight into the honeybee host–microbiome interaction.
Стилі APA, Harvard, Vancouver, ISO та ін.
23

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.

Повний текст джерела
Анотація:
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.
Стилі APA, Harvard, Vancouver, ISO та ін.
24

Tonomura, Shuichi, and Bibek Gyanwali. "Cerebral microbleeds in vascular dementia from clinical aspects to host-microbial interaction." Neurochemistry International 148 (September 2021): 105073. http://dx.doi.org/10.1016/j.neuint.2021.105073.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
25

Moreiras, Hugo, Mafalda Lopes‐da‐Silva, Miguel C. Seabra, and Duarte C. Barral. "Melanin processing by keratinocytes: A non‐microbial type of host‐pathogen interaction?" Traffic 20, no. 4 (March 15, 2019): 301–4. http://dx.doi.org/10.1111/tra.12638.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
26

Ahn, Jeonghyun, and Glen N. Barber. "STING signaling and host defense against microbial infection." Experimental & Molecular Medicine 51, no. 12 (December 2019): 1–10. http://dx.doi.org/10.1038/s12276-019-0333-0.

Повний текст джерела
Анотація:
AbstractThe first line of host defense against infectious agents involves activation of innate immune signaling pathways that recognize specific pathogen-associated molecular patterns (PAMPs). Key triggers of innate immune signaling are now known to include microbial-specific nucleic acid, which is rapidly detected in the cytosol of the cell. For example, RIG-I-like receptors (RLRs) have evolved to detect viral RNA species and to activate the production of host defense molecules and cytokines that stimulate adaptive immune responses. In addition, host defense countermeasures, including the production of type I interferons (IFNs), can also be triggered by microbial DNA from bacteria, viruses and perhaps parasites and are regulated by the cytosolic sensor, stimulator of interferon genes (STING). STING-dependent signaling is initiated by cyclic dinucleotides (CDNs) generated by intracellular bacteria following infection. CDNs can also be synthesized by a cellular synthase, cGAS, following interaction with invasive cytosolic self-DNA or microbial DNA species. The importance of STING signaling in host defense is evident since numerous pathogens have developed strategies to prevent STING function. Here, we review the relevance of STING-controlled innate immune signaling in host defense against pathogen invasion, including microbial endeavors to subvert this critical process.
Стилі APA, Harvard, Vancouver, ISO та ін.
27

Thakur, Aneesh, Heidi Mikkelsen, and Gregers Jungersen. "Intracellular Pathogens: Host Immunity and Microbial Persistence Strategies." Journal of Immunology Research 2019 (April 14, 2019): 1–24. http://dx.doi.org/10.1155/2019/1356540.

Повний текст джерела
Анотація:
Infectious diseases caused by pathogens including viruses, bacteria, fungi, and parasites are ranked as the second leading cause of death worldwide by the World Health Organization. Despite tremendous improvements in global public health since 1950, a number of challenges remain to either prevent or eradicate infectious diseases. Many pathogens can cause acute infections that are effectively cleared by the host immunity, but a subcategory of these pathogens called “intracellular pathogens” can establish persistent and sometimes lifelong infections. Several of these intracellular pathogens manage to evade the host immune monitoring and cause disease by replicating inside the host cells. These pathogens have evolved diverse immune escape strategies and overcome immune responses by residing and multiplying inside host immune cells, primarily macrophages. While these intracellular pathogens that cause persistent infections are phylogenetically diverse and engage in diverse immune evasion and persistence strategies, they share common pathogen type-specific mechanisms during host-pathogen interaction inside host cells. Likewise, the host immune system is also equipped with a diverse range of effector functions to fight against the establishment of pathogen persistence and subsequent host damage. This article provides an overview of the immune effector functions used by the host to counter pathogens and various persistence strategies used by intracellular pathogens to counter host immunity, which enables their extended period of colonization in the host. The improved understanding of persistent intracellular pathogen-derived infections will contribute to develop improved disease diagnostics, therapeutics, and prophylactics.
Стилі APA, Harvard, Vancouver, ISO та ін.
28

McAllister, T. A., K. A. Beauchemin, A. Y. Alazzeh, J. Baah, R. M. Teather, and K. Stanford. "Review: The use of direct fed microbials to mitigate pathogens and enhance production in cattle." Canadian Journal of Animal Science 91, no. 2 (June 2011): 193–211. http://dx.doi.org/10.4141/cjas10047.

Повний текст джерела
Анотація:
McAllister, T. A., Beauchemin, K. A., Alazzeh, A. Y., Baah, J., Teather, R. M. and Stanford, K. 2011. Review: The use of direct fed microbials to mitigate pathogens and enhance production in cattle. Can. J. Anim. Sci. 91: 193–211. Direct-fed microbials (DFM) have been employed in ruminant production for over 30 yr. Originally, DFM were used primarily in young ruminants to accelerate establishment of the intestinal microflora involved in feed digestion and to promote gut health. Further advancements led to more sophisticated mixtures of DFM that are targeted at improving fiber digestion and preventing ruminal acidosis in mature cattle. Through these outcomes on fiber digestion/rumen health, second-generation DFM have also resulted in improvements in milk yield, growth and feed efficiency of cattle, but results have been inconsistent. More recently, there has been an emphasis on the development of DFM that exhibit activity in cattle against potentially zoonotic pathogens such as Escherichia coli O157:H7, Salmonella spp. and Staphylococcus aureus. Regulatory requirements have limited the microbial species within DFM products to organisms that are generally recognized as safe, such as lactic acid-producing bacteria (e.g., Lactobacillus and Enterococcus spp.), fungi (e.g., Aspergillus oryzae), or yeast (e.g., Saccharomyces cerevisiae). Direct-fed microbials of rumen origin, involving lactate-utilizing species (e.g., Megasphaera elsdenii, Selenomonas ruminantium, Propionibacterium spp.) and plant cell wall-degrading isolates of Butyrivibrio fibrisolvens have also been explored, but have not been commercially used. Development of DFM that are efficacious over a wide range of ruminant production systems remains challenging because[0] comprehensive knowledge of microbial ecology is lacking. Few studies have employed molecular techniques to study in detail the interaction of DFM with native microbial communities or the ruminant host. Advancements in the metagenomics of microbial communities and the genomics of microbial–host interactions may enable DFM to be formulated to improve production and promote health, responses that are presently often achieved through the use of antimicrobials in cattle.
Стилі APA, Harvard, Vancouver, ISO та ін.
29

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.

Повний текст джерела
Анотація:
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.
Стилі APA, Harvard, Vancouver, ISO та ін.
30

Ghosh, Asit Ranjan. "Appraisal of Microbial Evolution to Commensalism and Pathogenicity in Humans." Clinical Medicine Insights: Gastroenterology 6 (January 2013): CGast.S11858. http://dx.doi.org/10.4137/cgast.s11858.

Повний текст джерела
Анотація:
The human body is host to a number of microbes occurring in various forms of host-microbe associations, such as commensals, mutualists, pathogens and opportunistic symbionts. While this association with microbes in certain cases is beneficial to the host, in many other cases it seems to offer no evident benefit or motive. The emergence and re-emergence of newer varieties of infectious diseases with causative agents being strains that were once living in the human system makes it necessary to study the environment and the dynamics under which this host microbe relationship thrives. The present discussion examines this interaction while tracing the origins of this association, and attempts to hypothesize a possible framework of selective pressures that could have lead microbes to inhabit mammalian host systems.
Стилі APA, Harvard, Vancouver, ISO та ін.
31

Portet, Anaïs, Eve Toulza, Ana Lokmer, Camille Huot, David Duval, Richard Galinier, and Benjamin Gourbal. "Experimental Infection of the Biomphalaria glabrata Vector Snail by Schistosoma mansoni Parasites Drives Snail Microbiota Dysbiosis." Microorganisms 9, no. 5 (May 18, 2021): 1084. http://dx.doi.org/10.3390/microorganisms9051084.

Повний текст джерела
Анотація:
Host-parasite interaction can result in a strong alteration of the host-associated microbiota. This dysbiosis can affect the fitness of the host; can modify pathogen interaction and the outcome of diseases. Biomphalaria glabrata is the snail intermediate host of the trematode Schistosoma mansoni, the agent of human schistosomiasis, causing hundreds of thousands of deaths every year. Here, we present the first study of the snail bacterial microbiota in response to Schistosoma infection. We examined the interplay between B. glabrata, S. mansoni and host microbiota. Snails were infected and the microbiota composition was analysed by 16S rDNA amplicon sequencing approach. We demonstrated that the microbial composition of water did not affect the microbiota composition. Then, we characterised the Biomphalaria bacterial microbiota at the individual scale in both naive and infected snails. Sympatric and allopatric strains of parasites were used for infections and re-infections to analyse the modification or dysbiosis of snail microbiota in different host-parasite co-evolutionary contexts. Concomitantly, using RNAseq, we investigated the link between bacterial microbiota dysbiosis and snail anti-microbial peptide immune response. This work paves the way for a better understanding of snail/schistosome interaction and should have critical consequences in terms of snail control strategies for fighting schistosomiasis disease in the field.
Стилі APA, Harvard, Vancouver, ISO та ін.
32

Muhamadali, Howbeer, Catherine L. Winder, Warwick B. Dunn, and Royston Goodacre. "Unlocking the secrets of the microbiome: exploring the dynamic microbial interplay with humans through metabolomics and their manipulation for synthetic biology applications." Biochemical Journal 480, no. 12 (June 28, 2023): 891–908. http://dx.doi.org/10.1042/bcj20210534.

Повний текст джерела
Анотація:
Metabolomics is a powerful research discovery tool with the potential to measure hundreds to low thousands of metabolites. In this review, we discuss the application of GC–MS and LC–MS in discovery-based metabolomics research, we define metabolomics workflows and we highlight considerations that need to be addressed in order to generate robust and reproducible data. We stress that metabolomics is now routinely applied across the biological sciences to study microbiomes from relatively simple microbial systems to their complex interactions within consortia in the host and the environment and highlight this in a range of biological species and mammalian systems including humans. However, challenges do still exist that need to be overcome to maximise the potential for metabolomics to help us understanding biological systems. To demonstrate the potential of the approach we discuss the application of metabolomics in two broad research areas: (1) synthetic biology to increase the production of high-value fine chemicals and reduction in secondary by-products and (2) gut microbial interaction with the human host. While burgeoning in importance, the latter is still in its infancy and will benefit from the development of tools to detangle host–gut-microbial interactions and their impact on human health and diseases.
Стилі APA, Harvard, Vancouver, ISO та ін.
33

Hu, S., A. R. Bourgonje, R. Gacesa, B. H. Jansen, A. Bangma, I. Hidding, E. A. M. Festen, et al. "P086 Mucosal microbiota modulate host intestinal immune signatures in Inflammatory Bowel Disease." Journal of Crohn's and Colitis 16, Supplement_1 (January 1, 2022): i185—i186. http://dx.doi.org/10.1093/ecco-jcc/jjab232.215.

Повний текст джерела
Анотація:
Abstract Background Host intestinal immune gene signatures and microbial dysregulations expose potential mechanisms in the pathogenesis of inflammatory bowel diseases (IBD). Profiling of mucosa-attached microbiota allows the understanding of locally present microbial communities and their immediate impact on the host. This study evaluated interactions between host mucosal gene expression and intestinal mucosa-attached microbiota in IBD. Methods Intestinal mucosal bulk RNA-sequencing data was combined with mucosal 16S rRNA gene sequencing data from 696 intestinal biopsies derived from 337 patients with IBD (181 with Crohn’s disease [CD] and 156 with ulcerative colitis [UC]) and 16 non-IBD controls. Hierarchical all-against-all associations testing (HAllA) was used to assess factors affecting host gene expressions and microbiota. Mucosal cell enrichments were predicted by deconvolution. Linear mixed interaction models were used to investigate host-microbiota interactions, adjusting for age, sex, BMI and batch effects. Variation explanation analysis was performed by Lasso regression. Results In total, 15,934 intestinal genes and 113 microbial taxa were identified and included in subsequent analyses. Host intestinal gene expressions were characterized by tissue- and inflammation-specificity, whereas intraindividual variability of the mucosal microbiota dominated over disease location and inflammation effects. We observed forty associations between the mucosal expression of genes and the abundance of specific microbes independent of dysbiosis (FDR<0.05). Examples include a positive association between aryl hydrocarbon receptor (AHR) and Bifidobacterium, and a negative association between interleukin 18 receptor 1 (IL18R1) and Lachnoclostridium. Furthermore, 112 gene-microbiota interactions changed in patients with microbial dysbiosis compared to non-dysbiosis (FDR<0.05). These interactions were enriched in immune-related and extracellular matrix organization pathways. For example, the IL1R1 gene was positively associated with Collinsella abundance in non-dysbiotic patients, whereas an inverse association was observed in high dysbiosis. Finally, the presence of mucosal microbial taxa explained up to 10% of the variation in cell type enrichment, affecting epithelial cells, macrophages and regulatory T-cells. Conclusion Interactions between host intestinal gene expressions and mucosa-attached microbiota are disrupted in patients with IBD. Furthermore, mucosal microbiota are highly personalized and potentially contribute to intestinal cell type alterations. Our study unravels key immune-mediated molecular pathways and relevant bacteria in intestinal tissue, which may guide drug development and precision medicine in IBD.
Стилі APA, Harvard, Vancouver, ISO та ін.
34

Martínez Arbas, Susana, Shaman Narayanasamy, Malte Herold, Laura A. Lebrun, Michael R. Hoopmann, Sujun Li, Tony J. Lam, et al. "Roles of bacteriophages, plasmids and CRISPR immunity in microbial community dynamics revealed using time-series integrated meta-omics." Nature Microbiology 6, no. 1 (November 2, 2020): 123–35. http://dx.doi.org/10.1038/s41564-020-00794-8.

Повний текст джерела
Анотація:
AbstractViruses and plasmids (invasive mobile genetic elements (iMGEs)) have important roles in shaping microbial communities, but their dynamic interactions with CRISPR-based immunity remain unresolved. We analysed generation-resolved iMGE–host dynamics spanning one and a half years in a microbial consortium from a biological wastewater treatment plant using integrated meta-omics. We identified 31 bacterial metagenome-assembled genomes encoding complete CRISPR–Cas systems and their corresponding iMGEs. CRISPR-targeted plasmids outnumbered their bacteriophage counterparts by at least fivefold, highlighting the importance of CRISPR-mediated defence against plasmids. Linear modelling of our time-series data revealed that the variation in plasmid abundance over time explained more of the observed community dynamics than phages. Community-scale CRISPR-based plasmid–host and phage–host interaction networks revealed an increase in CRISPR-mediated interactions coinciding with a decrease in the dominant ‘Candidatus Microthrix parvicella’ population. Protospacers were enriched in sequences targeting genes involved in the transmission of iMGEs. Understanding the factors shaping the fitness of specific populations is necessary to devise control strategies for undesirable species and to predict or explain community-wide phenotypes.
Стилі APA, Harvard, Vancouver, ISO та ін.
35

Diaz, Juan Manuel, Pravil Pokharel, and Alma Lilian Guerrero-Barrera. "Cellular Microbiology: Indispensable Tool to Dissect Host Pathogen Interaction." Current Trends in Biomedical Engineering & Biosciences 21, no. 1 (August 2, 2022): 01–04. http://dx.doi.org/10.19080/ctbeb.2022.21.556051.

Повний текст джерела
Анотація:
The threat of the spread of new or old infectious diseases are always a concern for human civilization. The emergence of coronavirus disease (COVID-19) and its dissemination around the world has demonstrated the risk to human health and the economy. So, it is essential to study a complete understanding of the host-pathogen interaction for the development of the disease. The field of cellular microbiology can help in the identification and characterization of different virulence factors produced by pathogens during each step of the infection process by combining techniques and approaches of classic cell biology and microbiology. Currently, the dynamic interaction of the host-pathogen relationship has been enhanced due to the cell culture, in vivo methods, in addition to microbial genomics, bioinformatics techniques, and in silico prediction methods to understand these diseases for the development of prevention methods, diagnosis and treatment worldwide.
Стилі APA, Harvard, Vancouver, ISO та ін.
36

Wommack, K. Eric, and Rita R. Colwell. "Virioplankton: Viruses in Aquatic Ecosystems." Microbiology and Molecular Biology Reviews 64, no. 1 (March 1, 2000): 69–114. http://dx.doi.org/10.1128/mmbr.64.1.69-114.2000.

Повний текст джерела
Анотація:
SUMMARY The discovery that viruses may be the most abundant organisms in natural waters, surpassing the number of bacteria by an order of magnitude, has inspired a resurgence of interest in viruses in the aquatic environment. Surprisingly little was known of the interaction of viruses and their hosts in nature. In the decade since the reports of extraordinarily large virus populations were published, enumeration of viruses in aquatic environments has demonstrated that the virioplankton are dynamic components of the plankton, changing dramatically in number with geographical location and season. The evidence to date suggests that virioplankton communities are composed principally of bacteriophages and, to a lesser extent, eukaryotic algal viruses. The influence of viral infection and lysis on bacterial and phytoplankton host communities was measurable after new methods were developed and prior knowledge of bacteriophage biology was incorporated into concepts of parasite and host community interactions. The new methods have yielded data showing that viral infection can have a significant impact on bacteria and unicellular algae populations and supporting the hypothesis that viruses play a significant role in microbial food webs. Besides predation limiting bacteria and phytoplankton populations, the specific nature of virus-host interaction raises the intriguing possibility that viral infection influences the structure and diversity of aquatic microbial communities. Novel applications of molecular genetic techniques have provided good evidence that viral infection can significantly influence the composition and diversity of aquatic microbial communities.
Стилі APA, Harvard, Vancouver, ISO та ін.
37

Li, Xiang-Yi, Tim Lachnit, Sebastian Fraune, Thomas C. G. Bosch, Arne Traulsen, and Michael Sieber. "Temperate phages as self-replicating weapons in bacterial competition." Journal of The Royal Society Interface 14, no. 137 (December 2017): 20170563. http://dx.doi.org/10.1098/rsif.2017.0563.

Повний текст джерела
Анотація:
Microbial communities are accompanied by a diverse array of viruses. Through infections of abundant microbes, these viruses have the potential to mediate competition within the community, effectively weakening competitive interactions and promoting coexistence. This is of particular relevance for host-associated microbial communities, because the diversity of the microbiota has been linked to host health and functioning. Here, we study the interaction between two key members of the microbiota of the freshwater metazoan Hydra vulgaris . The two commensal bacteria Curvibacter sp. and Duganella sp. protect their host from fungal infections, but only if both of them are present. Coexistence of the two bacteria is thus beneficial for Hydra . Intriguingly, Duganella sp. appears to be the superior competitor in vitro due to its higher growth rate when both bacteria are grown separately, but in co-culture the outcome of competition depends on the relative initial abundances of the two species. The presence of an inducible prophage in the Curvibacter sp. genome, which is able to lytically infect Duganella sp., led us to hypothesize that the phage modulates the interaction between these two key members of the Hydra microbiota. Using a mathematical model, we show that the interplay of the lysogenic life cycle of the Curvibacter phage and the lytic life cycle on Duganella sp. can explain the observed complex competitive interaction between the two bacteria. Our results highlight the importance of taking lysogeny into account for understanding microbe–virus interactions and show the complex role phages can play in promoting coexistence of their bacterial hosts.
Стилі APA, Harvard, Vancouver, ISO та ін.
38

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.

Повний текст джерела
Анотація:
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.
Стилі APA, Harvard, Vancouver, ISO та ін.
39

Matsuzaki, K. "Why and how are peptide-lipid interactions utilized for self defence?" Biochemical Society Transactions 29, no. 4 (August 1, 2001): 598–601. http://dx.doi.org/10.1042/bst0290598.

Повний текст джерела
Анотація:
Animals defend themselves against invading pathogenic micro-organisms by utilizing cationic anti-microbial peptides, which rapidly kill various micro-organisms without exerting toxicity against the host. Physicochemical peptide-lipid interactions provide attractive mechanisms for innate immunity. Many of these peptides form amphipathic secondary structures (α-helices and β-sheets) which can selectively interact with anionic bacterial membranes by electrostatic interaction. Rapid, peptide-induced membrane permeabilization is an effective mechanism of anti-microbial action. Magainin 2 from frog skin forms a dynamic peptide-lipid supramolecular-complex pore that allows mutually coupled transmembrane transport of ions and lipids. The peptide molecule is internalized upon the disintegration of the pore. Several anti-microbial peptides are known to work synergistically.
Стилі APA, Harvard, Vancouver, ISO та ін.
40

Scannapieco, Frank A. "Saliva-Bacterium Interactions in Oral Microbial Ecology." Critical Reviews in Oral Biology & Medicine 5, no. 3 (September 1994): 203–48. http://dx.doi.org/10.1177/10454411940050030201.

Повний текст джерела
Анотація:
Saliva is thought to have a significant impact on the colonization of microorganisms in the oral cavity. Salivary components may participate in this process by one of four general mechanisms: binding to microorganisms to facilitate their clearance from the oral cavity, serving as receptors in oral pellicles for microbial adhesion to host surfaces, inhibiting microbial growth or mediating microbial killing, and serving as microbial nutritional substrates. This article reviews information pertinent to the molecular interaction of salivary components with bacteria (primarily the oral streptococci and Actinomyces) and explores the implications of these interactions for oral bacterial colonization and dental plaque formation. Knowledge of the molecular mechanisms controlling bacterial colonization of the oral cavity may suggest methods to prevent not only dental plaque formation but also serious medical infections that may follow microbial colonization of the oral cavity.
Стилі APA, Harvard, Vancouver, ISO та ін.
41

Inman, R. D. "Immunogenetic aspects of host immune response." Canadian Journal of Microbiology 34, no. 3 (March 1, 1988): 319–22. http://dx.doi.org/10.1139/m88-058.

Повний текст джерела
Анотація:
The central role of histocompatibility leukocyte antigens (HLA) class II molecules in antigen presentation has received great attention in recent years, yet class I molecules have been defined as primarily functioning as a restriction element for cytotoxic T cell killing of virus-infected cells. Extensive clinical evidence, however, indicates that the HLA class I genes are strongly associated with nonseptic complications of enteric and genitourinary bacterial infections. Ninety percent of patients with Reiter's syndrome and reactive arthritis are positive for HLA-B27, yet the mechanism of disease susceptibility conferred by this gene remains obscure. Hypotheses concerning this interaction include (i) class I antigens functioning as receptors for microbial antigens; (ii) class I antigens expressing determinants that cross-react with microbial antigens; and (iii) class I genes controlling immunoregulatory functions that dictate qualitative differences in immune response to pathogenic organisms. These hypotheses await formal testing and hold great promise for understanding immunogenetic control of immune responses in general.
Стилі APA, Harvard, Vancouver, ISO та ін.
42

Casadevall, Arturo, and Liise-anne Pirofski. "What Is a Host? Incorporating the Microbiota into the Damage-Response Framework: TABLE 1." Infection and Immunity 83, no. 1 (November 10, 2014): 2–7. http://dx.doi.org/10.1128/iai.02627-14.

Повний текст джерела
Анотація:
Since proof of the germ theory of disease in the late 19th century, a major focus of the fields of microbiology and infectious diseases has been to seek differences between pathogenic and nonpathogenic microbes and the role that the host plays in microbial pathogenesis. Remarkably, despite the increasing recognition that host immunity plays a role in microbial pathogenesis, there has been little discussion about what constitutes a host. Historically, hosts have been viewed in the context of their fitness or immunological status and characterized by adjectives such as immune, immunocompetent, immunosuppressed, immunocompromised, or immunologically impaired. However, in recent years it has become apparent that the microbiota has profound effects on host homeostasis and susceptibility to microbial diseases in addition to its effects on host immunity. This raises the question of how to incorporate the microbiota into defining a host. This definitional problem is further complicated because neither host nor microbial properties are adequate to predict the outcome of host-microbe interaction because this outcome exhibits emergent properties. In this essay, we revisit the damage-response framework (DRF) of microbial pathogenesis and demonstrate how it can incorporate the rapidly accumulating information being generated by the microbiome revolution. We use the tenets of the DRF to put forth the following definition of a host: a host is an entity that houses an associated microbiome/microbiota and interacts with microbes such that the outcome results in damage, benefit, or indifference, thus resulting in the states of symbiosis, colonization, commensalism, latency, and disease.
Стилі APA, Harvard, Vancouver, ISO та ін.
43

Byrd, Warren C., Sarah Schwartz-Baxter, Jim Carlson, Silvana Barros, Steven Offenbacher, and Sompop Bencharit. "Role of salivary and candidal proteins in denture stomatitis: an exploratory proteomic analysis." Mol. BioSyst. 10, no. 9 (2014): 2299–304. http://dx.doi.org/10.1039/c4mb00185k.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
44

Pathak, Parul, Vineet Kumar Rai, Hasan CAN, Sandeep Kumar Singh, Dharmendra Kumar, Nikunj Bhardwaj, Rajib Roychowdhury, et al. "Plant-Endophyte Interaction during Biotic Stress Management." Plants 11, no. 17 (August 25, 2022): 2203. http://dx.doi.org/10.3390/plants11172203.

Повний текст джерела
Анотація:
Plants interact with diverse microbial communities and share complex relationships with each other. The intimate association between microbes and their host mutually benefit each other and provide stability against various biotic and abiotic stresses to plants. Endophytes are heterogeneous groups of microbes that live inside the host tissue without showing any apparent sign of infection. However, their functional attributes such as nutrient acquisition, phytohormone modulation, synthesis of bioactive compounds, and antioxidant enzymes of endophytes are similar to the other rhizospheric microorganisms. Nevertheless, their higher colonization efficacy and stability against abiotic stress make them superior to other microorganisms. In recent studies, the potential role of endophytes in bioprospecting has been broadly reported. However, the molecular aspect of host–endophyte interactions is still unclear. In this study, we have briefly discussed the endophyte biology, colonization efficacy and diversity pattern of endophytes. In addition, it also summarizes the molecular aspect of plant–endophyte interaction in biotic stress management.
Стилі APA, Harvard, Vancouver, ISO та ін.
45

Li, Zexin, Donald Pan, Guangshan Wei, Weiling Pi, Chuwen Zhang, Jiang-Hai Wang, Yongyi Peng, et al. "Deep sea sediments associated with cold seeps are a subsurface reservoir of viral diversity." ISME Journal 15, no. 8 (March 1, 2021): 2366–78. http://dx.doi.org/10.1038/s41396-021-00932-y.

Повний текст джерела
Анотація:
AbstractIn marine ecosystems, viruses exert control on the composition and metabolism of microbial communities, influencing overall biogeochemical cycling. Deep sea sediments associated with cold seeps are known to host taxonomically diverse microbial communities, but little is known about viruses infecting these microorganisms. Here, we probed metagenomes from seven geographically diverse cold seeps across global oceans to assess viral diversity, virus–host interaction, and virus-encoded auxiliary metabolic genes (AMGs). Gene-sharing network comparisons with viruses inhabiting other ecosystems reveal that cold seep sediments harbour considerable unexplored viral diversity. Most cold seep viruses display high degrees of endemism with seep fluid flux being one of the main drivers of viral community composition. In silico predictions linked 14.2% of the viruses to microbial host populations with many belonging to poorly understood candidate bacterial and archaeal phyla. Lysis was predicted to be a predominant viral lifestyle based on lineage-specific virus/host abundance ratios. Metabolic predictions of prokaryotic host genomes and viral AMGs suggest that viruses influence microbial hydrocarbon biodegradation at cold seeps, as well as other carbon, sulfur and nitrogen cycling via virus-induced mortality and/or metabolic augmentation. Overall, these findings reveal the global diversity and biogeography of cold seep viruses and indicate how viruses may manipulate seep microbial ecology and biogeochemistry.
Стилі APA, Harvard, Vancouver, ISO та ін.
46

Claus, Sandrine P., Sandrine L. Ellero, Bernard Berger, Lutz Krause, Anne Bruttin, Jérôme Molina, Alain Paris, et al. "Colonization-Induced Host-Gut Microbial Metabolic Interaction." mBio 2, no. 2 (March 1, 2011). http://dx.doi.org/10.1128/mbio.00271-10.

Повний текст джерела
Анотація:
ABSTRACT The gut microbiota enhances the host’s metabolic capacity for processing nutrients and drugs and modulate the activities of multiple pathways in a variety of organ systems. We have probed the systemic metabolic adaptation to gut colonization for 20 days following exposure of axenic mice (n = 35) to a typical environmental microbial background using high-resolution 1H nuclear magnetic resonance (NMR) spectroscopy to analyze urine, plasma, liver, kidney, and colon (5 time points) metabolic profiles. Acquisition of the gut microbiota was associated with rapid increase in body weight (4%) over the first 5 days of colonization with parallel changes in multiple pathways in all compartments analyzed. The colonization process stimulated glycogenesis in the liver prior to triggering increases in hepatic triglyceride synthesis. These changes were associated with modifications of hepatic Cyp8b1 expression and the subsequent alteration of bile acid metabolites, including taurocholate and tauromuricholate, which are essential regulators of lipid absorption. Expression and activity of major drug-metabolizing enzymes (Cyp3a11 and Cyp2c29) were also significantly stimulated. Remarkably, statistical modeling of the interactions between hepatic metabolic profiles and microbial composition analyzed by 16S rRNA gene pyrosequencing revealed strong associations of the Coriobacteriaceae family with both the hepatic triglyceride, glucose, and glycogen levels and the metabolism of xenobiotics. These data demonstrate the importance of microbial activity in metabolic phenotype development, indicating that microbiota manipulation is a useful tool for beneficially modulating xenobiotic metabolism and pharmacokinetics in personalized health care. IMPORTANCE Gut bacteria have been associated with various essential biological functions in humans such as energy harvest and regulation of blood pressure. Furthermore, gut microbial colonization occurs after birth in parallel with other critical processes such as immune and cognitive development. Thus, it is essential to understand the bidirectional interaction between the host metabolism and its symbionts. Here, we describe the first evidence of an in vivo association between a family of bacteria and hepatic lipid metabolism. These results provide new insights into the fundamental mechanisms that regulate host-gut microbiota interactions and are thus of wide interest to microbiological, nutrition, metabolic, systems biology, and pharmaceutical research communities. This work will also contribute to developing novel strategies in the alteration of host-gut microbiota relationships which can in turn beneficially modulate the host metabolism.
Стилі APA, Harvard, Vancouver, ISO та ін.
47

Ahmed, EM. "Microbial Endocrinology: Interaction of the Microbial Hormones with the Host." Biomedical Journal of Scientific & Technical Research 24, no. 2 (January 6, 2020). http://dx.doi.org/10.26717/bjstr.2020.24.004015.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
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.

Повний текст джерела
Анотація:
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.
Стилі APA, Harvard, Vancouver, ISO та ін.
49

Wan, Tingting, Yalong Wang, Kaixin He, and Shu Zhu. "Microbial sensing in the intestine." Protein & Cell, May 16, 2023. http://dx.doi.org/10.1093/procel/pwad028.

Повний текст джерела
Анотація:
Abstract The gut microbiota plays a key role in host health and disease, particularly through their interactions with the immune system. Intestinal homeostasis is dependent on the symbiotic relationships between the host and the diverse gut microbiota, which is influenced by the highly co-evolved immune-microbiota interactions. The first step of the interaction between the host and the gut microbiota is the sensing of the gut microbes by the host immune system. In this review, we describe the cells of the host immune system and the proteins that sense the components and metabolites of the gut microbes. We further highlight the essential roles of pattern recognition receptors (PRRs), the G protein coupled receptors (GPCRs), aryl hydrocarbon receptor (AHR) and the nuclear receptors expressed in the intestinal epithelial cells (IECs) and the intestine-resident immune cells. We also discuss the mechanisms by which the disruption of microbial sensing because of genetic or environmental factors causes human diseases such as the inflammatory bowel disease (IBD).
Стилі APA, Harvard, Vancouver, ISO та ін.
50

Hernández-Rocha, Cristian, Krzysztof Borowski, Williams Turpin, Melissa Filice, Shadi Nayeri, Juan Antonio Raygoza Garay, Joanne M. Stempak, and Mark S. Silverberg. "Integrative analysis of colonic biopsies from inflammatory bowel disease patients identifies an interaction between microbial bile-acid inducible gene abundance and human Angiopoietin-like 4 gene expression." Journal of Crohn's and Colitis, June 2, 2021. http://dx.doi.org/10.1093/ecco-jcc/jjab096.

Повний текст джерела
Анотація:
Abstract Background and Aims Microbial derived-bile acids can modulate host gene expression, and their fecal abundance is decreased in active inflammatory bowel disease (IBD). We analyzed the impact of endoscopic inflammation on microbial genes involved in bile acid biotransformation, and their interaction with host transcriptome in the intestinal mucosa of IBD patients. Methods Endoscopic mucosal biopsies were collected from non-inflamed and inflamed terminal ileum, ascending and sigmoid colon of IBD patients. Prediction of imputed metagenome functional content from 16S rRNA profile and real-time qPCR were utilized to assess microbial bile acid biotransformation gene abundance, and RNA-seq was used for host transcriptome analysis. Linear regression and partial Spearman correlation accounting for age, sex and IBD type were used to assess the association between microbial genes, inflammation and host transcriptomics in each biopsy location. A Bayesian network (BN) analysis was fitted to infer the direction of interactions between IBD traits, microbial and host genes. Results Inferred microbial gene pathway involved in secondary bile acid biosynthesis (ko00121 pathway) was depleted in inflamed terminal ileum of IBD patients compared to non-inflamed tissue. In non-inflamed sigmoid colon, the relative abundance of bile acid-inducible (baiCD) microbial genes was positively correlated with the host Angiopoietin-like 4 (Angptl4) gene expression. The BN analysis suggests that the microbial baiCD gene abundance could affect Angptl4 expression, and this interaction appears to be lost in the presence of inflammation. Conclusions Endoscopic inflammation affects the abundance of crucial microbial bile acid-metabolizing genes and their interaction with Angptl4 in intestinal mucosa of IBD patients.
Стилі APA, Harvard, Vancouver, ISO та ін.
Ми пропонуємо знижки на всі преміум-плани для авторів, чиї праці увійшли до тематичних добірок літератури. Зв'яжіться з нами, щоб отримати унікальний промокод!

До бібліографії