Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 422
Filter
1.
BMC Biol ; 22(1): 224, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379910

ABSTRACT

BACKGROUND: Nematodes are the most abundant metazoans in marine sediments, many of which are bacterivores; however, how habitat bacteria affect physiological outcomes in marine nematodes remains largely unknown.  RESULTS: Here, we used a Litoditis marina inbred line to assess how native bacteria modulate host nematode physiology. We characterized seasonal dynamic bacterial compositions in L. marina habitats and examined the impacts of 448 habitat bacteria isolates on L. marina development, then focused on HQbiome with 73 native bacteria, of which we generated 72 whole genomes sequences. Unexpectedly, we found that the effects of marine native bacteria on the development of L. marina and its terrestrial relative Caenorhabditis elegans were significantly positively correlated. Next, we reconstructed bacterial metabolic networks and identified several bacterial metabolic pathways positively correlated with L. marina development (e.g., ubiquinol and heme b biosynthesis), while pyridoxal 5'-phosphate biosynthesis pathway was negatively associated. Through single metabolite supplementation, we verified CoQ10, heme b, acetyl-CoA, and acetaldehyde promoted L. marina development, while vitamin B6 attenuated growth. Notably, we found that only four development correlated metabolic pathways were shared between L. marina and C. elegans. Furthermore, we identified two bacterial metabolic pathways correlated with L. marina lifespan, while a distinct one in C. elegans. Strikingly, we found that glycerol supplementation significantly extended L. marina but not C. elegans longevity. Moreover, we comparatively demonstrated the distinct gut microbiota characteristics and their effects on L. marina and C. elegans physiology. CONCLUSIONS: Given that both bacteria and marine nematodes are dominant taxa in sedimentary ecosystems, the resource presented here will provide novel insights to identify mechanisms underpinning how habitat bacteria affect nematode biology in a more natural context. Our integrative approach will provide a microbe-nematodes framework for microbiome mediated effects on host animal fitness.


Subject(s)
Caenorhabditis elegans , Microbiota , Animals , Microbiota/physiology , Caenorhabditis elegans/physiology , Caenorhabditis elegans/microbiology , Nematoda/physiology , Nematoda/microbiology , Bacteria/genetics , Bacteria/classification , Bacteria/isolation & purification , Bacteria/metabolism , Ecosystem
2.
Microbiol Spectr ; : e0086124, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39373496

ABSTRACT

Understanding factors influencing the composition and maintenance of beneficial host-associated microbial communities is central to understanding their ecological, evolutionary, and health consequences for hosts. Host immunity is often implicated as a regulator of these microbiota, but immunity may also play a disruptive role, with responses to infection perturbing beneficial communities. Such effects may be more prominent from innate immune responses, with more rapid-acting and often non-specific components, compared to adaptive responses. We investigated how upregulation of antibacterial immunity in the bumble bee Bombus impatiens affects its core gut microbiota, testing the hypothesis that immunity-induced perturbation impacts the microbiota structure. Freshly emerged adult bees were fed a microbiota inoculum before receiving a non-pathogenic immune stimulation injection. We quantified microbial communities using 16S rRNA amplicon sequencing and targeted quantitative PCR. Coarse community membership shows apparent robustness, but we find that immune stimulation alters the abundance of two core community members, Gilliamella and Snodgrassella. Moreover, a positive association in communities between these bacteria is perturbed following a Gram-negative challenge. The observed changes in the gut microbial community are suggestive of immune response-induced dysbiosis, linking ecological interactions across levels between hosts, their pathogens, and their beneficial gut microbiota. The potential for collateral perturbation of the natural gut microbiota following an innate immune response may contribute to immune costs, shaping the evolutionary optimization of immune investment depending on the ecological context. IMPORTANCE: Our work demonstrates how innate immunity may influence the host-associated microbiota. While previous work has demonstrated the role of adaptive immunity in regulating the microbiota, we show that stimulation of an innate immune response in bumble bees may disrupt the native gut microbial community by shifting individual abundances of some members and pairwise associations. This work builds upon previous work in bumble bees demonstrating factors determining microbe colonization of hosts and microbiota membership, implicating immune response-induced changes as a factor shaping these important gut communities. While some microbiota members appear unaffected, changes in others and the community overall suggests that collateral perturbation of the native gut microbiota upon an innate immune response may serve as an additional selective pressure that shapes the evolution of host innate immunity.

3.
mBio ; : e0238724, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39254316

ABSTRACT

The microbiome plays a vital role in human health, with changes in its composition impacting various aspects of the body. Posttranslational modification (PTM) regulates protein activity by attaching chemical groups to amino acids in an enzymatic or non-enzymatic manner. PTMs offer fast and dynamic regulation of protein expression and can be influenced by specific dietary components that induce PTM events in gut microbiomes and their hosts. PTMs on microbiome proteins have been found to contribute to host-microbe interactions. For example, in Escherichia coli, S-sulfhydration of tryptophanase regulates uremic toxin production and chronic kidney disease in mice. On a broader microbial scale, the microbiomes of patients with inflammatory bowel disease exhibit distinct PTM patterns in their metaproteomes. Moreover, pathogens and commensals can alter host PTM profiles through protein secretion and diet-regulated metabolic shifts. The emerging field of metaPTMomics focuses on understanding PTM profiles in the microbiota, their association with lifestyle factors like diet, and their functional effects on host-microbe interactions.

4.
J Adv Res ; 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39233003

ABSTRACT

INTRODUCTION: Host-microbe interactions are important to human health and ecosystems globally, so elucidating the complex host-microbe interactions and associated protein expressions drives the need to develop sensitive and accurate biochemical techniques. Current proteomics techniques reveal information from the point of view of either the host or microbe, but do not provide data on the corresponding partner. Moreover, it remains challenging to simultaneously study host-microbe proteomes that reflect the direct competition between host and microbe. This raises the need to develop a dual-species proteomics method for host-microbe interactions. OBJECTIVES: We aim to establish a forward + reverse Stable Isotope Labeling with Amino acids in Cell culture (SILAC) proteomics approach to simultaneously label and quantify newly-expressed proteins of host and microbe without physical isolation, for investigating mechanisms in direct host-microbe interactions. METHODS: Using Caenorhabditis elegans-Pseudomonas aeruginosa infection model as proof-of-concept, we employed SILAC proteomics and molecular pathway analysis to characterize the differentially-expressed microbial and host proteins. We then used molecular docking and chemical characterization to identify chemical inhibitors that intercept host-microbe interactions and eliminate microbial infection. RESULTS: Based on our proteomics results, we studied the iron competition between pathogen iron scavenger and host iron uptake protein, where P. aeruginosa upregulated pyoverdine synthesis protein (PvdA) (fold-change of 5.2313) and secreted pyoverdine, and C. elegans expressed ferritin (FTN-2) (fold-change of 3.4057). Targeted intervention of iron competition was achieved using Galangin, a ginger-derived phytochemical that inhibited pyoverdine production and biofilm formation in P. aeruginosa. The Galangin-ciprofloxacin combinatorial therapy could eliminate P. aeruginosa biofilms in a fish wound infection model, and enabled animal survival. CONCLUSION: Our work provides a novel SILAC-based proteomics method that can simultaneously evaluate host and microbe proteomes, with future applications in higher host organisms and other microbial species. It also provides insights into the mechanisms dictating host-microbe interactions, offering novel strategies for anti-infective therapy.

5.
Sheng Wu Gong Cheng Xue Bao ; 40(9): 2916-2933, 2024 Sep 25.
Article in Chinese | MEDLINE | ID: mdl-39319715

ABSTRACT

The human gut is a complex ecosystem harboring rich microbes that play a key role in the nutrient absorption, drug metabolism, and immune responses. With the continuous development of microfluidics and organ-on-a-chip, gut-on-a-chip has become a powerful tool for modeling host-microbe interactions. The chip is able to mimic the complex physiological environment of the human gut in vitro, providing a unique platform for studying host-microbe interactions. Firstly, we introduce the physiological characteristics of the human gut. Secondly, we comprehensively summarize the advantages of the microfluidic chip in vitro recapitulating the intestinal system by integrating microenvironmental factors, such as complex cell components, dynamic fluids, oxygen gradients, and mechanical mechanics. Thirdly, we expound the key performance indicators for evaluating the construction performance of gut-on-a-chip. In addition, we review the progress of gut-on-a-chip models in the research on gut microecology, disease modeling, and drug evaluation. Finally, we highlight the challenges and prospects in the applications of the emerging technology. The above is summarized with a view to informing the application of gut-on-a-chip for indepth studies of gut microbe-host interactions.


Subject(s)
Gastrointestinal Microbiome , Lab-On-A-Chip Devices , Humans , Host Microbial Interactions , Gastrointestinal Tract/microbiology , Intestines/microbiology
6.
J Microbiol ; 62(9): 709-725, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39240507

ABSTRACT

The importance of the complex interplay between the microbiome and mucosal immunity, particularly within the respiratory tract, has gained significant attention due to its potential implications for the severity and progression of lung diseases. Therefore, this review summarizes the specific interactions through which the respiratory tract-specific microbiome influences mucosal immunity and ultimately impacts respiratory health. Furthermore, we discuss how the microbiome affects mucosal immunity, considering tissue-specific variations, and its capacity in respiratory diseases containing asthma, chronic obstructive pulmonary disease, and lung cancer. Additionally, we investigate the external factors which affect the relationship between respiratory microbiome and mucosal immune responses. By exploring these intricate interactions, this review provides valuable insights into the potential for microbiome-based interventions to modulate mucosal immunity and alleviate the severity of respiratory diseases.


Subject(s)
Disease Progression , Immunity, Mucosal , Microbiota , Humans , Microbiota/immunology , Asthma/immunology , Asthma/microbiology , Respiratory System/microbiology , Respiratory System/immunology , Animals , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/microbiology , Lung Neoplasms/immunology , Lung Neoplasms/microbiology
7.
APMIS ; 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39270740

ABSTRACT

Several microbiome studies have recently demonstrated microbial dysbiosis in various chronic inflammatory skin diseases, and it is considered an important role in the pathogenesis. Although the role of skin dysbiosis in inflammatory skin diseases is debatable, the local microenvironment is considered essential concerning compositional changes and functional alterations of the skin microbiota. Indeed, various local nutrients (e.g., lipids), pH values, water, oxygen, and antimicrobial peptides may affect the level of skin dysbiosis in these skin diseases. In particular, in atopic dermatitis and hidradenitis suppurativa, significant changes in skin dysbiosis have been associated with local aberrant host immune changes. In this review, the potential pathogenic crosstalk between the host and the microbiota is reviewed in relation to the physical, chemical, and biological microenvironments of various chronic inflammatory skin diseases.

8.
mSphere ; 9(9): e0026224, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39158277

ABSTRACT

The study of the fecal microbiota is crucial for unraveling the pathways through which gut symbionts are acquired and transmitted. While stable gut microbial communities are essential for honey bee health, their modes of acquisition and transmission are yet to be confirmed. The gut of honey bees is colonized by symbiotic bacteria within 5 days after emergence from their wax cells as adults. Few studies have suggested that bees could be colonized in part via contact with fecal matter in the hive. However, the composition of the fecal microbiota is still unknown. It is particularly unclear whether all bacterial species can be found viable in the feces and can therefore be transmitted to newborn nestmates. Using 16S rRNA gene amplicon sequencing, we revealed that the composition of the honey bee fecal microbiota is strikingly similar to the microbiota of entire guts. We found that fecal transplantation resulted in gut microbial communities similar to those obtained from feeding gut homogenates. Our study shows that fecal sampling and transplantation are viable tools for the non-invasive analysis of bacterial community composition and host-microbe interactions. It also implies that contact of young bees with fecal matter in the hive is a plausible route for gut microbiota acquisition. IMPORTANCE: Honey bees are crucial pollinators for many crops and wildflowers. They are also powerful models for studying microbiome-host interactions. However, current methods rely on gut tissue disruption to analyze microbiota composition and use gut homogenates to inoculate microbiota-deprived bees. Here, we provide two new and non-invasive approaches that will open doors to longitudinal studies: fecal sampling and transplantation. Furthermore, our findings provide insights into gut microbiota transmission in social insects by showing that ingestion of fecal matter can result in gut microbiota acquisition.


Subject(s)
Bacteria , Fecal Microbiota Transplantation , Feces , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Animals , Bees/microbiology , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Host Microbial Interactions , DNA, Bacterial/genetics , Symbiosis
9.
Gut Microbes ; 16(1): 2385117, 2024.
Article in English | MEDLINE | ID: mdl-39120056

ABSTRACT

Autism spectrum disorders (ASD) are highly heritable, heterogeneous neurodevelopmental disorders characterized by clinical presentation of atypical social, communicative, and repetitive behaviors. Over the past 25 years, hundreds of ASD risk genes have been identified. Many converge on key molecular pathways, from translational control to those regulating synaptic structure and function. Despite these advances, therapeutic approaches remain elusive. Emerging data unearthing the relationship between genetics, microbes, and immunity in ASD suggest an integrative physiology approach could be paramount to delivering therapeutic breakthroughs. Indeed, the advent of large-scale multi-OMIC data acquisition, analysis, and interpretation is yielding an increasingly mechanistic understanding of ASD and underlying risk factors, revealing how genetic susceptibility interacts with microbial genetics, metabolism, epigenetic (re)programming, and immunity to influence neurodevelopment and behavioral outcomes. It is now possible to foresee exciting advancements in the treatment of some forms of ASD that could markedly improve quality of life and productivity for autistic individuals. Here, we highlight recent work revealing how gene X maternal exposome interactions influence risk for ASD, with emphasis on the intrauterine environment and fetal neurodevelopment, host-microbe interactions, and the evolving therapeutic landscape for ASD.


Subject(s)
Autism Spectrum Disorder , Humans , Autism Spectrum Disorder/microbiology , Autism Spectrum Disorder/etiology , Female , Pregnancy , Exposome , Gastrointestinal Microbiome , Risk Factors , Genetic Predisposition to Disease , Animals , Autistic Disorder/etiology , Autistic Disorder/microbiology
10.
Elife ; 132024 Aug 27.
Article in English | MEDLINE | ID: mdl-39190452

ABSTRACT

Host-microbe interactions are virtually bidirectional, but how the host affects their microbiome is poorly understood. Here, we report that the host is a critical modulator to regulate the lifestyle switch and pathogenicity heterogeneity of the opportunistic pathogens Serratia marcescens utilizing the Drosophila and bacterium model system. First, we find that Drosophila larvae efficiently outcompete S. marcescens and typically drive a bacterial switch from pathogenicity to commensalism toward the fly. Furthermore, Drosophila larvae reshape the transcriptomic and metabolic profiles of S. marcescens characterized by a lifestyle switch. More importantly, the host alters pathogenicity and heterogeneity of S. marcescens in the single-cell resolution. Finally, we find that larvae-derived AMPs are required to recapitulate the response of S. marcescens to larvae. Altogether, our findings provide an insight into the pivotal roles of the host in harnessing the life history and heterogeneity of symbiotic bacterial cells, advancing knowledge of the reciprocal relationships between the host and pathogen.


Subject(s)
Drosophila melanogaster , Host-Pathogen Interactions , Larva , Serratia marcescens , Animals , Serratia marcescens/pathogenicity , Serratia marcescens/genetics , Serratia marcescens/physiology , Larva/microbiology , Drosophila melanogaster/microbiology , Single-Cell Analysis , Symbiosis , Drosophila/microbiology , Virulence/genetics
11.
Ecol Evol ; 14(7): e11699, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39041011

ABSTRACT

Microorganisms are important associates of insect and arthropod species. Insect-associated microbes, including bacteria, fungi, and viruses, can drastically impact host physiology, ecology, and fitness, while many microbes still have no known role. Over the past decade, we have increased our knowledge of the taxonomic composition and functional roles of insect-associated microbiomes and viromes. There has been a more recent shift toward examining the complexity of microbial communities, including how they vary in response to different factors (e.g., host genome, microbial strain, environment, and time), and the consequences of this variation for the host and the wider ecological community. We provide an overview of insect-microbe interactions, the variety of associated microbial functions, and the evolutionary ecology of these relationships. We explore the influence of the environment and the interactive effects of insects and their microbiomes across trophic levels. Additionally, we discuss the potential for subsequent synergistic and reciprocal impacts on the associated microbiomes, ecological interactions, and communities. Lastly, we discuss some potential avenues for the future of insect-microbe interactions that include the modification of existing microbial symbionts as well as the construction of synthetic microbial communities.

12.
Ecol Evol ; 14(7): e11705, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38975267

ABSTRACT

Endosymbionts are widespread in arthropods, living in host cells with effects that extend from parasitic to mutualistic. Newly acquired endosymbionts tend to be parasitic, but vertical transmission favors coevolution toward mutualism, with hosts sometimes developing dependency. Endosymbionts negatively affecting host fitness may still spread by impacting host reproductive traits, referred to as reproductive "manipulation," although costs for hosts are often assumed rather than demonstrated. For cytoplasmic incompatibility (CI) that involves endosymbiont-mediated embryo death, theory predicts directional shifts away from "manipulation" toward reduced CI strength; moreover, CI-causing endosymbionts need to increase host fitness to initially spread. In nature, endosymbiont-host interactions and dynamics are complex, often depending on environmental conditions and evolutionary history. We advocate for capturing this complexity through appropriate datasets, rather than relying on terms like "manipulation." Such imprecision can lead to the misclassification of endosymbionts along the parasitism-mutualism continuum.

13.
Front Microbiol ; 15: 1434346, 2024.
Article in English | MEDLINE | ID: mdl-39050639

ABSTRACT

The study aimed to explore the similarities and differences in gut microorganisms and their functions in regulating body mass in Eothenomys miletus across different altitudes in the Hengduan Mountains when exposed to a high-fat diet. Eothenomys miletus specimens were gathered from Dali (DL) and Xianggelila (XGLL) in Yunnan Province, China, and categorized into control, high-fat (1 week of high-fat diet), and re-feeding groups (1 week of high-fat diet followed by 2 weeks of standard food). The analysis utilized 16S rRNA sequencing to assess the diversity and structure of intestinal microbial communities in E. miletus. The investigation focused on the impact of high-fat diet consumption and different altitudes on gut microbial diversity, structure, and physiological markers. Results revealed that a high-fat diet influenced the beta diversity of gut microorganisms in E. miletus, leading to variations in microbial community structure between the two regions with different altitudes. High-fat food significantly affected body mass, white adipose tissue mass, triglycerides, and leptin levels, but not food intake. Specific intestinal microorganisms were observed in the high-fat groups, aiding in food digestion and being enriched in particular flora. In particular, microbial genera like Lactobacillus and Hylemonella were enriched in the high-fat group of DL. The enriched microbiota in the control group was associated with plant polysaccharide and cellulose decomposition. Following a high-fat diet, gut microbiota adapted to support lipid metabolism and energy supply, while upon re-feeding, the focus shifted back to cellulose digestion. These findings suggested that alterations in gut microbial composition, alongside physiological markers, play a vital role in adaptation of E. miletus to the diverse habitats of the Hengduan Mountains at varying altitudes.

14.
Fish Shellfish Immunol ; 152: 109788, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39053586

ABSTRACT

In the process of screening for probiotic strains, there are no clearly established bacterial phenotypic markers which could be used for the prediction of their in vivo mechanism of action. In this work, we demonstrate for the first time that Machine Learning (ML) methods can be used for accurately predicting the in vivo immunomodulatory activity of probiotic strains based on their cell surface phenotypic features using a snail host-microbe interaction model. A broad range of snail gut presumptive probiotics, including 240 new lactic acid bacterial strains (Lactobacillus, Leuconostoc, Lactococcus, and Enterococcus), were isolated and characterized based on their capacity to withstand snails' gastrointestinal defense barriers, such as the pedal mucus, gastric mucus, gastric juices, and acidic pH, in association with their cell surface hydrophobicity, autoaggregation, and biofilm formation ability. The implemented ML pipeline predicted with high accuracy (88 %) strains with a strong capacity to enhance chemotaxis and phagocytic activity of snails' hemolymph cells, while also revealed bacterial autoaggregation and cell surface hydrophobicity as the most important parameters that significantly affect host immune responses. The results show that ML approaches may be useful to derive a predictive understanding of host-probiotic interactions, while also highlighted the use of snails as an efficient animal model for screening presumptive probiotic strains in the light of their interaction with cellular innate immune responses.


Subject(s)
Machine Learning , Probiotics , Probiotics/pharmacology , Animals , Lactobacillales/physiology , Lactobacillales/immunology , Snails/immunology , Snails/microbiology , Helix, Snails/immunology , Helix, Snails/physiology , Immunity, Innate , Immunomodulation
15.
Fish Shellfish Immunol ; 152: 109797, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39084276

ABSTRACT

Bacterial intestinal inflammation is a common disease of yellow catfish (Pelteobagrus fulvidraco) in high-density aquaculture. Understanding the interactions between host and intestinal bacteria is helpful to intestinal inflammatory disease control. Here, we constructed a model of intestinal inflammation after Aeromonas hydrophila infection in yellow catfish, and characterized variations in gene expression and microbiome in the gut through high-throughput sequencing. Furthermore, host gene-microbiome interactions were identified. Histology observation showed disordered distribution of columnar epithelial cells and decrease of goblet cells in intestine. A total of 4741 genes showed differentially expression, mostly in comparisons between 12 hpi group with each other groups respectively, including control, 24 hpi and 48 hpi groups. These genes were enriched in immune-related pathways including the IL-17 signaling pathway, triggering strong inflammatory response at the invading stage within 12 h. Subsequently, the host strengthened energy consumption by activating carbohydrate and lipid metabolism pathways to repair the intestinal mucosal immune defense line. In addition, fish with A. hydrophila infection show decreased richness of gut microbial, reduced relative abundance of probiotics including Akkermansia, and elevated pathogenic bacteria such as Plesimonas. An integrative analysis identified A. hydrophila-related genes, such as il22 and stat3, for which expression level is close associated with the shift of A. hydrophila-related bacteria relative abundance, such as Akkermansia and Cetobacterium. Aside from picturing the variations of intestine gene expression and mucosal microbiome of yellow catfish coping with A. hydrophila infection, our study probed the underlying host-microbe interactions in A. hydrophila infection induced intestinal inflammatory, providing new insights for disease control in aquaculture.


Subject(s)
Aeromonas hydrophila , Catfishes , Fish Diseases , Gastrointestinal Microbiome , Gram-Negative Bacterial Infections , Animals , Aeromonas hydrophila/physiology , Catfishes/immunology , Catfishes/genetics , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Fish Diseases/immunology , Fish Diseases/microbiology
16.
Microbiol Resour Announc ; 13(8): e0033024, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39037312

ABSTRACT

We present the whole-genome sequence of Enterobacter hormaechei (previously Enterobacter cloacae) obtained from long and short reads. It is a dominant gut symbiont of the notorious crop pest Plutella xylostella, highly prevalent in lepidopteran midguts and a useful model for the evolution of resistance to antimicrobials.

17.
Microbiome Res Rep ; 3(2): 20, 2024.
Article in English | MEDLINE | ID: mdl-38841412

ABSTRACT

Cell culture is a powerful technique for the investigation of molecular mechanisms fundamental to health and disease in a diverse array of organisms. Cell lines offer several advantages, namely their simplistic approach and high degree of reproducibility. One field where cell culture has proven particularly useful is the study of the microbiome, where cell culture has led to the illumination of microbial influences on host immunity, nutrition, and physiology. Thus far, researchers have focused cell culture work predominantly on humans, but the growing field of insect microbiome research stands to benefit greatly from its application. Insects constitute one of Earth's most diverse and ancient life forms and, just as with humans, possess microbiomes with great significance to their health. Insects, which play critical roles in supporting food security and ecological stability, are facing increasing threats from agricultural intensification, climate change, and pesticide use. As the microbiome is closely tied to host health, gaining a more robust understanding is of increasing importance. In this review, we assert that the cultivation and utilization of insect gut cell lines in microbiome research will bridge critical knowledge gaps essential for informing insect management practices in a world under pressure.

18.
Methods Mol Biol ; 2820: 127-137, 2024.
Article in English | MEDLINE | ID: mdl-38941020

ABSTRACT

Intestinal fungi are a fundamental component of the gut microbiome and play important roles in mammalian host biology. At the same time, the contribution of gut fungi to host health and disease remains understudied due to their low abundance. In that respect, gnotobiotic animals with defined microbial populations of reduced complexity represent a well-suited model system that highlights the effects of low abundant gut fungi on host physiology and other members of the microbial community. In this chapter, a label-free quantitative metaproteomic approach for the characterization of simplified microbial communities in gnotobiotic mice is presented. The model allows for exploring various research questions on the role of gut fungi in disease pathogenesis, microbial ecosystem maturation, or host-microbiome crosstalk.


Subject(s)
Fungi , Gastrointestinal Microbiome , Germ-Free Life , Proteomics , Animals , Mice , Proteomics/methods , Fungi/metabolism
19.
J Microbiol Biol Educ ; 25(2): e0004624, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-38888313

ABSTRACT

The current and ongoing challenges brought on by climate change will require future scientists who have hands-on experience using advanced molecular techniques, can work with large data sets, and can make correlations between metadata and microbial diversity. A course-embedded research project can prepare students to answer complex research questions that might help plants adapt to climate change. The project described herein uses plants as a host to study the impact of climate change-induced drought on host-microbe interactions through next-generation DNA sequencing and analysis using a command-line program. Specifically, the project studies the impact of simulated drought on the rhizosphere microbiome of Fast Plants rapid cycling Brassica rapa using inexpensive greenhouse supplies and 16S rRNA V3/V4 Illumina sequencing. Data analysis is performed with the freely accessible Python-based microbiome bioinformatics platform QIIME 2.

20.
Immunol Rev ; 325(1): 107-130, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38864582

ABSTRACT

Autoimmune (or rheumatic) diseases are increasing in prevalence but selecting the best therapy for each patient proceeds in trial-and-error fashion. This strategy can lead to ineffective therapy resulting in irreversible damage and suffering; thus, there is a need to bring the promise of precision medicine to patients with autoimmune disease. While host factors partially determine the therapeutic response to immunosuppressive drugs, these are not routinely used to tailor therapy. Thus, non-host factors likely contribute. Here, we consider the impact of the human gut microbiome in the treatment of autoimmunity. We propose that the gut microbiome can be manipulated to improve therapy and to derive greater benefit from existing therapies. We focus on the mechanisms by which the human gut microbiome impacts treatment response, provide a framework to interrogate these mechanisms, review a case study of a widely-used anti-rheumatic drug, and discuss challenges with studying multiple complex systems: the microbiome, the human immune system, and autoimmune disease. We consider open questions that remain in the field and speculate on the future of drug-microbiome-autoimmune disease interactions. Finally, we present a blue-sky vision for how the microbiome can be used to bring the promise of precision medicine to patients with rheumatic disease.


Subject(s)
Autoimmune Diseases , Gastrointestinal Microbiome , Precision Medicine , Humans , Gastrointestinal Microbiome/immunology , Autoimmune Diseases/therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/microbiology , Animals , Autoimmunity , Immunosuppressive Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL