Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.369
Filter
1.
Front Cell Infect Microbiol ; 14: 1407051, 2024.
Article in English | MEDLINE | ID: mdl-38947127

ABSTRACT

The Cecum is a key site for cellulose digestion in nutrient metabolism of intestine, but its mechanisms of microbial and gene interactions has not been fully elucidated during pathogenesis of obesity. Therefore, the cecum tissues of the New Zealand rabbits and their contents between the high-fat diet-induced group (Ob) and control group (Co) were collected and analyzed using multi-omics. The metagenomic analysis indicated that the relative abundances of Corallococcus_sp._CAG:1435 and Flavobacteriales bacterium species were significantly lower, while those of Akkermansia glycaniphila, Clostridium_sp._CAG:793, Mycoplasma_sp._CAG:776, Mycoplasma_sp._CAG:472, Clostridium_sp._CAG:609, Akkermansia_sp._KLE1605, Clostridium_sp._CAG:508, and Firmicutes_bacterium_CAG:460 species were significantly higher in the Ob as compared to those in Co. Transcriptomic sequencing results showed that the differentially upregulated genes were mainly enriched in pathways, including calcium signaling pathway, PI3K-Akt signaling pathway, and Wnt signaling pathway, while the differentially downregulated genes were mainly enriched in pathways of NF-kappaB signaling pathway and T cell receptor signaling pathway. The comparative analysis of metabolites showed that the glycine, serine, and threonine metabolism and cysteine and methionine metabolism were the important metabolic pathways between the two groups. The combined analysis showed that CAMK1, IGFBP6, and IGFBP4 genes were highly correlated with Clostridium_sp._CAG:793, and Akkermansia_glycaniphila species. Thus, the preliminary study elucidated the microbial and gene interactions in cecum of obese rabbit and provided a basis for further studies in intestinal intervention for human obesity.


Subject(s)
Cecum , Diet, High-Fat , Gastrointestinal Microbiome , Obesity , Animals , Rabbits , Diet, High-Fat/adverse effects , Cecum/microbiology , Cecum/metabolism , Obesity/metabolism , Obesity/microbiology , Host Microbial Interactions , Metagenomics , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Bacteria/isolation & purification , Gene Regulatory Networks , Male , Gene Expression Profiling
2.
Environ Microbiol ; 26(7): e16670, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38952172

ABSTRACT

The influence of environmental factors on the interactions between phages and bacteria, particularly single-stranded DNA (ssDNA) phages, has been largely unexplored. In this study, we used Finnlakevirus FLiP, the first known ssDNA phage species with a lipid membrane, as our model phage. We examined the infectivity of FLiP with three Flavobacterium host strains, B330, B167 and B114. We discovered that FLiP infection is contingent on the host strain and conditions such as temperature and bacterial growth phase. FLiP can infect its hosts across a wide temperature range, but optimal phage replication varies with each host. We uncovered some unique aspects of phage infectivity: FLiP has limited infectivity in liquid-suspended cells, but it improves when cells are surface-attached. Moreover, FLiP infects stationary phase B167 and B114 cells more rapidly and efficiently than exponentially growing cells, a pattern not observed with the B330 host. We also present the first experimental evidence of endolysin function in ssDNA phages. The activity of FLiP's lytic enzymes was found to be condition-dependent. Our findings underscore the importance of studying phage ecology in contexts that are relevant to the environment, as both the host and the surrounding conditions can significantly alter the outcome of phage-host interactions.


Subject(s)
Bacteriophages , DNA, Single-Stranded , Flavobacterium , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Bacteriophages/genetics , Bacteriophages/physiology , Flavobacterium/virology , Flavobacterium/growth & development , Flavobacterium/genetics , Host Microbial Interactions , Endopeptidases/metabolism , Endopeptidases/genetics , Virus Replication , Temperature
3.
Rev Med Virol ; 34(4): e2567, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38937135

ABSTRACT

Human T-lymphotropic virus type-1 (HTLV-1) was the first discovered human oncogenic retrovirus, the etiological agent of two serious diseases have been identified as adult T-cell leukaemia/lymphoma malignancy and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), a debilitating chronic neuro-myelopathy. Despite more than 40 years of molecular, histopathological and immunological studies on HTLV-1-associated diseases, the virulence and pathogenicity of this virus are yet to be clarified. The reason why the majority of HTLV-1-infected individuals (∼95%) remain asymptomatic carriers is still unclear. The deterioration of the immune system towards oncogenicity and autoimmunity makes HTLV-1 a natural probe for the study of malignancy and neuro-inflammatory diseases. Additionally, its slow worldwide spreading has prompted public health authorities and researchers, as urged by the WHO, to focus on eradicating HTLV-1. In contrast, neither an effective therapy nor a protective vaccine has been introduced. This comprehensive review focused on the most relevant studies of the neuro-inflammatory propensity of HTLV-1-induced HAM/TSP. Such an emphasis on the virus-host interactions in the HAM/TSP pathogenesis will be critically discussed epigenetically. The findings may shed light on future research venues in designing and developing proper HTLV-1 therapeutics.


Subject(s)
HTLV-I Infections , Human T-lymphotropic virus 1 , Paraparesis, Tropical Spastic , Humans , Human T-lymphotropic virus 1/pathogenicity , Human T-lymphotropic virus 1/physiology , Paraparesis, Tropical Spastic/virology , Paraparesis, Tropical Spastic/immunology , HTLV-I Infections/virology , HTLV-I Infections/immunology , HTLV-I Infections/complications , Host-Pathogen Interactions/immunology , Animals , Host Microbial Interactions/immunology
4.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928375

ABSTRACT

Microbes constitute the most prevalent life form on Earth, yet their remarkable diversity remains mostly unrecognized. Microbial diversity in vertebrate models presents a significant challenge for investigating host-microbiome interactions. The model organism Caenorhabditis elegans has many advantages for delineating the effects of host genetics on microbial composition. In the wild, the C. elegans gut contains various microbial species, while in the laboratory it is usually a host for a single bacterial species. There is a potential host-microbe interaction between microbial metabolites, drugs, and C. elegans phenotypes. This mini-review aims to summarize the current understanding regarding the microbiome in C. elegans. Examples using C. elegans to study host-microbe-metabolite interactions are discussed.


Subject(s)
Caenorhabditis elegans , Animals , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/genetics , Gastrointestinal Microbiome , Models, Animal , Microbiota , Host Microbial Interactions , Bacteria/genetics , Bacteria/classification , Bacteria/metabolism
5.
Front Cell Infect Microbiol ; 14: 1407261, 2024.
Article in English | MEDLINE | ID: mdl-38846354

ABSTRACT

SARS-CoV-2 is the causative virus of the devastating COVID-19 pandemic that results in an unparalleled global health and economic crisis. Despite unprecedented scientific efforts and therapeutic interventions, the fight against COVID-19 continues as the rapid emergence of different SARS-CoV-2 variants of concern and the increasing challenge of long COVID-19, raising a vast demand to understand the pathomechanisms of COVID-19 and its long-term sequelae and develop therapeutic strategies beyond the virus per se. Notably, in addition to the virus itself, the replication cycle of SARS-CoV-2 and clinical severity of COVID-19 is also governed by host factors. In this review, we therefore comprehensively overview the replication cycle and pathogenesis of SARS-CoV-2 from the perspective of host factors and host-virus interactions. We sequentially outline the pathological implications of molecular interactions between host factors and SARS-CoV-2 in multi-organ and multi-system long COVID-19, and summarize current therapeutic strategies and agents targeting host factors for treating these diseases. This knowledge would be key for the identification of new pathophysiological aspects and mechanisms, and the development of actionable therapeutic targets and strategies for tackling COVID-19 and its sequelae.


Subject(s)
COVID-19 , Host-Pathogen Interactions , SARS-CoV-2 , Virus Replication , Humans , COVID-19/virology , SARS-CoV-2/pathogenicity , Antiviral Agents/therapeutic use , Host Microbial Interactions
6.
Nat Commun ; 15(1): 4708, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830853

ABSTRACT

Critical illness can significantly alter the composition and function of the human microbiome, but few studies have examined these changes over time. Here, we conduct a comprehensive analysis of the oral, lung, and gut microbiota in 479 mechanically ventilated patients (223 females, 256 males) with acute respiratory failure. We use advanced DNA sequencing technologies, including Illumina amplicon sequencing (utilizing 16S and ITS rRNA genes for bacteria and fungi, respectively, in all sample types) and Nanopore metagenomics for lung microbiota. Our results reveal a progressive dysbiosis in all three body compartments, characterized by a reduction in microbial diversity, a decrease in beneficial anaerobes, and an increase in pathogens. We find that clinical factors, such as chronic obstructive pulmonary disease, immunosuppression, and antibiotic exposure, are associated with specific patterns of dysbiosis. Interestingly, unsupervised clustering of lung microbiota diversity and composition by 16S independently predicted survival and performed better than traditional clinical and host-response predictors. These observations are validated in two separate cohorts of COVID-19 patients, highlighting the potential of lung microbiota as valuable prognostic biomarkers in critical care. Understanding these microbiome changes during critical illness points to new opportunities for microbiota-targeted precision medicine interventions.


Subject(s)
COVID-19 , Dysbiosis , Gastrointestinal Microbiome , Lung , Microbiota , Humans , Female , Male , Dysbiosis/microbiology , Middle Aged , Lung/microbiology , COVID-19/microbiology , COVID-19/virology , Aged , Microbiota/genetics , Gastrointestinal Microbiome/genetics , Host Microbial Interactions/genetics , Longitudinal Studies , RNA, Ribosomal, 16S/genetics , Respiratory Insufficiency/microbiology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Adult , Respiration, Artificial , Bacteria/genetics , Bacteria/classification , Bacteria/isolation & purification , Critical Illness , Metagenomics/methods
7.
Cell Host Microbe ; 32(6): 820-836, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38870899

ABSTRACT

Microbial communities that colonize the human gastrointestinal (GI) tract defend against pathogens through a mechanism known as colonization resistance (CR). Advances in technologies such as next-generation sequencing, gnotobiotic mouse models, and bacterial cultivation have enhanced our understanding of the underlying mechanisms and the intricate microbial interactions involved in CR. Rather than being attributed to specific microbial clades, CR is now understood to arise from a dynamic interplay between microbes and the host and is shaped by metabolic, immune, and environmental factors. This evolving perspective underscores the significance of contextual factors, encompassing microbiome composition and host conditions, in determining CR. This review highlights recent research that has shifted its focus toward elucidating how these factors interact to either promote or impede enteric infections. It further discusses future research directions to unravel the complex relationship between host, microbiota, and environmental determinants in safeguarding against GI infections to promote human health.


Subject(s)
Gastrointestinal Microbiome , Humans , Animals , Mice , Host Microbial Interactions , Gastrointestinal Tract/microbiology , Bacteria/genetics , Bacteria/classification , Host-Pathogen Interactions , Germ-Free Life , Microbial Interactions
8.
Cell Host Microbe ; 32(6): 786-793, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38870896

ABSTRACT

Survival strategies of human-associated microbes to drug exposure have been mainly studied in the context of bona fide pathogens exposed to antibiotics. Less well understood are the survival strategies of non-pathogenic microbes and host-associated commensal communities to the variety of drugs and xenobiotics to which humans are exposed. The lifestyle of microbial commensals within complex communities offers a variety of ways to adapt to different drug-induced stresses. Here, we review the responses and survival strategies employed by gut commensals when exposed to drugs-antibiotics and non-antibiotics-at the individual and community level. We also discuss the factors influencing the recovery and establishment of a new community structure following drug exposure. These survival strategies are key to the stability and resilience of the gut microbiome, ultimately influencing the overall health and well-being of the host.


Subject(s)
Anti-Bacterial Agents , Bacteria , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/drug effects , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Xenobiotics/pharmacology , Symbiosis , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/drug effects , Drug Resistance, Bacterial , Host Microbial Interactions/drug effects
9.
NPJ Biofilms Microbiomes ; 10(1): 47, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898089

ABSTRACT

Throughout the life span of a host, bifidobacteria have shown superior colonization and glycan abilities. Complex glycans, such as human milk oligosaccharides and plant glycans, that reach the colon are directly internalized by the transport system of bifidobacteria, cleaved into simple structures by extracellular glycosyl hydrolase, and transported to cells for fermentation. The glycan utilization of bifidobacteria introduces cross-feeding activities between bifidobacterial strains and other microbiota, which are influenced by host nutrition and regulate gut homeostasis. This review discusses bifidobacterial glycan utilization strategies, focusing on the cross-feeding involved in bifidobacteria and its potential health benefits. Furthermore, the impact of cross-feeding on the gut trophic niche of bifidobacteria and host health is also highlighted. This review provides novel insights into the interactions between microbe-microbe and host-microbe.


Subject(s)
Bifidobacterium , Gastrointestinal Microbiome , Homeostasis , Polysaccharides , Humans , Bifidobacterium/metabolism , Bifidobacterium/physiology , Polysaccharides/metabolism , Host Microbial Interactions , Animals , Fermentation
10.
Gut Microbes ; 16(1): 2369337, 2024.
Article in English | MEDLINE | ID: mdl-38904092

ABSTRACT

The gut microbiota, comprising trillions of diverse microorganisms inhabiting the intestines of animals, forms a complex and indispensable ecosystem with profound implications for the host's well-being. Its functions include contributing to developing the host's immune response, aiding in nutrient digestion, synthesizing essential compounds, acting as a barrier against pathogen invasion, and influencing the development or regression of various pathologies. The dietary habits of the host directly impact this intricate community of gut microbes. Diet influences the composition and function of the gut microbiota through alterations in gene expression, enzymatic activity, and metabolome. While the impact of diet on gut ecology is well-established, the investigation into the relationship between dietary consumption and microbial genotypic diversity has been limited. This review provides an overview of the relationship between diet and gut microbiota, emphasizing the impact of host nutrition on both short- and long-term evolution in the mammalian gut. It is evident that the evolution of the gut microbiota occurs even on short timescales through the acquisition of novel mutations, within the gut bacteria of individual hosts. Consequently, we discuss the importance of considering alterations in bacterial genomic diversity when analyzing microbiota-dependent effects on host physiology. Future investigations into the various microbiota-related traits shall greatly benefit from a deeper understanding of commensal bacterial evolutionary adaptation.


Subject(s)
Bacteria , Diet , Gastrointestinal Microbiome , Symbiosis , Gastrointestinal Microbiome/physiology , Animals , Humans , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Biological Evolution , Host Microbial Interactions
11.
FEMS Microbiol Ecol ; 100(7)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38857884

ABSTRACT

Host-associated microbial communities are shaped by host migratory movements. These movements can have contrasting impacts on microbiota, and understanding such patterns can provide insight into the ecological processes that contribute to community diversity. Furthermore, long-distance movements to new environments are anticipated to occur with increasing frequency due to host distribution shifts resulting from climate change. Understanding how hosts transport their microbiota with them could be of importance when examining biological invasions. Although microbial community shifts are well-documented, the underlying mechanisms that lead to the restructuring of these communities remain relatively unexplored. Using literature and ecological simulations, we develop a framework to elucidate the major factors that lead to community change. We group host movements into two types-regular (repeated/cyclical migratory movements, as found in many birds and mammals) and irregular (stochastic/infrequent movements that do not occur on a cyclical basis, as found in many insects and plants). Ecological simulations and prior research suggest that movement type and frequency, alongside environmental exposure (e.g. internal/external microbiota) are key considerations for understanding movement-associated community changes. From our framework, we derive a series of testable hypotheses, and suggest means to test them, to facilitate future research into host movement and microbial community dynamics.


Subject(s)
Microbiota , Animals , Animal Migration , Biodiversity , Birds/microbiology , Climate Change , Host Microbial Interactions , Mammals/microbiology
12.
Toxins (Basel) ; 16(6)2024 May 24.
Article in English | MEDLINE | ID: mdl-38922136

ABSTRACT

Clostridioides difficile, a Gram-positive anaerobic bacterium, is the leading cause of hospital-acquired antibiotic-associated diarrhea worldwide. The severity of C. difficile infection (CDI) varies, ranging from mild diarrhea to life-threatening conditions such as pseudomembranous colitis and toxic megacolon. Central to the pathogenesis of the infection are toxins produced by C. difficile, with toxin A (TcdA) and toxin B (TcdB) as the main virulence factors. Additionally, some strains produce a third toxin known as C. difficile transferase (CDT). Toxins damage the colonic epithelium, initiating a cascade of cellular events that lead to inflammation, fluid secretion, and further tissue damage within the colon. Mechanistically, the toxins bind to cell surface receptors, internalize, and then inactivate GTPase proteins, disrupting the organization of the cytoskeleton and affecting various Rho-dependent cellular processes. This results in a loss of epithelial barrier functions and the induction of cell death. The third toxin, CDT, however, functions as a binary actin-ADP-ribosylating toxin, causing actin depolymerization and inducing the formation of microtubule-based protrusions. In this review, we summarize our current understanding of the interaction between C. difficile toxins and host cells, elucidating the functional consequences of their actions. Furthermore, we will outline how this knowledge forms the basis for developing innovative, toxin-based strategies for treating and preventing CDI.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Host Microbial Interactions , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Clostridium Infections/pathology , Gene Order , Inflammation/pathology , Humans , Animals
13.
Front Immunol ; 15: 1401320, 2024.
Article in English | MEDLINE | ID: mdl-38835769

ABSTRACT

Host-microbe interactions are complex and ever-changing, especially during infections, which can significantly impact human physiology in both health and disease by influencing metabolic and immune functions. Infections caused by pathogens such as bacteria, viruses, fungi, and parasites are the leading cause of global mortality. Microbes have evolved various immune evasion strategies to survive within their hosts, which presents a multifaceted challenge for detection. Intracellular microbes, in particular, target specific cell types for survival and replication and are influenced by factors such as functional roles, nutrient availability, immune evasion, and replication opportunities. Identifying intracellular microbes can be difficult because of the limitations of traditional culture-based methods. However, advancements in integrated host microbiome single-cell genomics and transcriptomics provide a promising basis for personalized treatment strategies. Understanding host-microbiota interactions at the cellular level may elucidate disease mechanisms and microbial pathogenesis, leading to targeted therapies. This article focuses on how intracellular microbes reside in specific cell types, modulating functions through persistence strategies to evade host immunity and prolong colonization. An improved understanding of the persistent intracellular microbe-induced differential disease outcomes can enhance diagnostics, therapeutics, and preventive measures.


Subject(s)
Genomics , Single-Cell Analysis , Humans , Genomics/methods , Animals , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Host Microbial Interactions/immunology , Host Microbial Interactions/genetics , Immune Evasion , Microbiota/immunology , Bacteria/genetics , Bacteria/immunology , Severity of Illness Index
14.
J R Soc Interface ; 21(215): 20230756, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38900957

ABSTRACT

The health and well-being of a host are deeply influenced by the interactions with its gut microbiota. Contrasted environmental conditions, such as diseases or dietary habits, play a pivotal role in modulating these interactions, impacting microbiota composition and functionality. Such conditions can also lead to transitions from beneficial to detrimental symbiosis, viewed as alternative stable states of the host-microbiota dialogue. This article introduces a novel mathematical model exploring host-microbiota interactions, integrating dynamics of the colonic epithelial crypt, microbial metabolic functions, inflammation sensitivity and colon flows in a transverse section. The model considers metabolic shifts in epithelial cells based on butyrate and hydrogen sulfide concentrations, innate immune pattern recognition receptor activation, microbial oxygen tolerance and the impact of antimicrobial peptides on the microbiota. Using the model, we demonstrated that a high-protein, low-fibre diet exacerbates detrimental interactions and compromises beneficial symbiotic resilience, underscoring a destabilizing effect towards an unhealthy state. Moreover, the proposed model provides essential insights into oxygen levels, fibre and protein breakdown, and basic mechanisms of innate immunity in the colon and offers a crucial understanding of factors influencing the colon environment.


Subject(s)
Gastrointestinal Microbiome , Models, Biological , Symbiosis , Humans , Gastrointestinal Microbiome/physiology , Symbiosis/physiology , Colon/metabolism , Colon/microbiology , Host Microbial Interactions/physiology , Host Microbial Interactions/immunology , Immunity, Innate
15.
ISME J ; 18(1)2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38896583

ABSTRACT

Probiotics have gained significant attention as a potential strategy to improve health by modulating host-microbe interactions, particularly in situations where the normal microbiota has been disrupted. However, evidence regarding their efficacy has been inconsistent, with considerable interindividual variability in response. We aimed to explore whether a common genetic variant that affects the production of mucosal α(1,2)-fucosylated glycans, present in around 20% of the population, could explain the observed interpersonal differences in the persistence of commonly used probiotics. Using a mouse model with varying α(1,2)-fucosylated glycans secretion (Fut2WT or Fut2KO), we examined the abundance and persistence of Bifidobacterium strains (infantis, breve, and bifidum). We observed significant differences in baseline gut microbiota characteristics between Fut2WT and Fut2KO littermates, with Fut2WT mice exhibiting enrichment of species able to utilize α(1,2)-fucosylated glycans. Following antibiotic exposure, only Fut2WT animals showed persistent engraftment of Bifidobacterium infantis, a strain able to internalize α(1,2)-fucosylated glycans, whereas B. breve and B. bifidum, which cannot internalize α(1,2)-fucosylated glycans, did not exhibit this difference. In mice with an intact commensal microbiota, the relationship between secretor status and B. infantis persistence was reversed, with Fut2KO animals showing greater persistence compared to Fut2WT. Our findings suggest that the interplay between a common genetic variation and antibiotic exposure plays a crucial role in determining the dynamics of B. infantis in the recipient gut, which could potentially contribute to the observed variation in response to this commonly used probiotic species.


Subject(s)
Anti-Bacterial Agents , Fucosyltransferases , Galactoside 2-alpha-L-fucosyltransferase , Gastrointestinal Microbiome , Probiotics , Animals , Mice , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Probiotics/administration & dosage , Anti-Bacterial Agents/pharmacology , Bifidobacterium longum subspecies infantis/genetics , Bifidobacterium longum subspecies infantis/metabolism , Polysaccharides/metabolism , Host Microbial Interactions , Mice, Inbred C57BL , Mice, Knockout , Bifidobacterium/genetics , Bifidobacterium/metabolism
16.
Retrovirology ; 21(1): 13, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898526

ABSTRACT

Retroviruses exploit host proteins to assemble and release virions from infected cells. Previously, most studies focused on interacting partners of retroviral Gag proteins that localize to the cytoplasm or plasma membrane. Given that several full-length Gag proteins have been found in the nucleus, identifying the Gag-nuclear interactome has high potential for novel findings involving previously unknown host processes. Here we systematically compared nuclear factors identified in published HIV-1 proteomic studies and performed our own mass spectrometry analysis using affinity-tagged HIV-1 and RSV Gag proteins mixed with nuclear extracts. We identified 57 nuclear proteins in common between HIV-1 and RSV Gag, and a set of nuclear proteins present in our analysis and ≥ 1 of the published HIV-1 datasets. Many proteins were associated with nuclear processes which could have functional consequences for viral replication, including transcription initiation/elongation/termination, RNA processing, splicing, and chromatin remodeling. Examples include facilitating chromatin remodeling to expose the integrated provirus, promoting expression of viral genes, repressing the transcription of antagonistic cellular genes, preventing splicing of viral RNA, altering splicing of cellular RNAs, or influencing viral or host RNA folding or RNA nuclear export. Many proteins in our pulldowns common to RSV and HIV-1 Gag are critical for transcription, including PolR2B, the second largest subunit of RNA polymerase II (RNAPII), and LEO1, a PAF1C complex member that regulates transcriptional elongation, supporting the possibility that Gag influences the host transcription profile to aid the virus. Through the interaction of RSV and HIV-1 Gag with splicing-related proteins CBLL1, HNRNPH3, TRA2B, PTBP1 and U2AF1, we speculate that Gag could enhance unspliced viral RNA production for translation and packaging. To validate one putative hit, we demonstrated an interaction of RSV Gag with Mediator complex member Med26, required for RNA polymerase II-mediated transcription. Although 57 host proteins interacted with both Gag proteins, unique host proteins belonging to each interactome dataset were identified. These results provide a strong premise for future functional studies to investigate roles for these nuclear host factors that may have shared functions in the biology of both retroviruses, as well as functions specific to RSV and HIV-1, given their distinctive hosts and molecular pathology.


Subject(s)
Gene Products, gag , HIV-1 , Humans , HIV-1/physiology , HIV-1/genetics , Gene Products, gag/metabolism , Gene Products, gag/genetics , Cell Nucleus/metabolism , Cell Nucleus/virology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , Rous sarcoma virus/physiology , Rous sarcoma virus/genetics , Proteomics , Host-Pathogen Interactions , Virus Replication , Host Microbial Interactions , Mass Spectrometry
17.
Gut Microbes ; 16(1): 2361660, 2024.
Article in English | MEDLINE | ID: mdl-38935764

ABSTRACT

The microbiota significantly impacts digestive epithelium functionality, especially in nutrient processing. Given the importance of iron for both the host and the microbiota, we hypothesized that host-microbiota interactions fluctuate with dietary iron levels. We compared germ-free (GF) and conventional mice (SPF) fed iron-containing (65 mg/Kg) or iron-depleted (<6 mg/Kg) diets. The efficacy of iron privation was validated by iron blood parameters. Ferritin and Dmt1, which represent cellular iron storage and transport respectively, were studied in tissues where they are abundant: the duodenum, liver and lung. When the mice were fed an iron-rich diet, the microbiota increased blood hemoglobin and hepcidin and the intestinal ferritin levels, suggesting that the microbiota helps iron storage. When iron was limiting, the microbiota inhibited the expression of the intestinal Dmt1 transporter, likely via the pathway triggered by Hif-2α. The microbiota assists the host in storing intestinal iron when it is abundant and competes with the host by inhibiting Dmt1 in conditions of iron scarcity. Comparison between duodenum, liver and lung indicates organ-specific responses to microbiota and iron availability. Iron depletion induced temporal changes in microbiota composition and activity, reduced α-diversity of microbiota, and led to Lactobacillaceae becoming particularly more abundant after 60 days of privation. By inoculating GF mice with a simplified bacterial mixture, we show that the iron-depleted host favors the gut fitness of Bifidobacterium longum.


Subject(s)
Cation Transport Proteins , Duodenum , Gastrointestinal Microbiome , Hepcidins , Iron, Dietary , Liver , Animals , Mice , Gastrointestinal Microbiome/physiology , Iron, Dietary/metabolism , Iron, Dietary/administration & dosage , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Liver/metabolism , Liver/microbiology , Duodenum/metabolism , Duodenum/microbiology , Hepcidins/metabolism , Ferritins/metabolism , Germ-Free Life , Host Microbial Interactions , Lung/microbiology , Lung/metabolism , Iron/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics , Bacteria/isolation & purification , Mice, Inbred C57BL , Hemoglobins/metabolism , Male
18.
Gut Microbes ; 16(1): 2351520, 2024.
Article in English | MEDLINE | ID: mdl-38717832

ABSTRACT

Links between the gut microbiota and human health have been supported throughout numerous studies, such as the development of neurological disease disorders. This link is referred to as the "microbiota-gut-brain axis" and is the focus of an emerging field of research. Microbial-derived metabolites and gut and neuro-immunological metabolites regulate this axis in health and many diseases. Indeed, assessing these signals, whether induced by microbial metabolites or neuro-immune mediators, could significantly increase our knowledge of the microbiota-gut-brain axis. However, this will require the development of appropriate techniques and potential models. Methods for studying the induced signals originating from the microbiota remain crucial in this field. This review discusses the methods and techniques available for studies of microbiota-gut-brain interactions. We highlight several much-debated elements of these methodologies, including the widely used in vivo and in vitro models, their implications, and perspectives in the field based on a systematic review of PubMed. Applications of various animal models (zebrafish, mouse, canine, rat, rabbit) to microbiota-gut-brain axis research with practical examples of in vitro methods and innovative approaches to studying gut-brain communications are highlighted. In particular, we extensively discuss the potential of "organ-on-a-chip" devices and their applications in this field. Overall, this review sheds light on the most widely used models and methods, guiding researchers in the rational choice of strategies for studies of microbiota-gut-brain interactions.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Host Microbial Interactions , Animals , Gastrointestinal Microbiome/physiology , Brain-Gut Axis/physiology , Humans , Brain/microbiology , Brain/metabolism , Brain/physiology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism , Models, Animal , Mice
19.
ISME J ; 18(1)2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38767866

ABSTRACT

Host-microbe interactions underlie the development and fitness of many macroorganisms, including bees. Whereas many social bees benefit from vertically transmitted gut bacteria, current data suggests that solitary bees, which comprise the vast majority of species diversity within bees, lack a highly specialized gut microbiome. Here, we examine the composition and abundance of bacteria and fungi throughout the complete life cycle of the ground-nesting solitary bee Anthophora bomboides standfordiana. In contrast to expectations, immature bee stages maintain a distinct core microbiome consisting of Actinobacterial genera (Streptomyces, Nocardiodes) and the fungus Moniliella spathulata. Dormant (diapausing) larval bees hosted the most abundant and distinctive bacteria and fungi, attaining 33 and 52 times their initial copy number, respectively. We tested two adaptive hypotheses regarding microbial functions for diapausing bees. First, using isolated bacteria and fungi, we found that Streptomyces from brood cells inhibited the growth of multiple pathogenic filamentous fungi, suggesting a role in pathogen protection during overwintering, when bees face high pathogen pressure. Second, sugar alcohol composition changed in tandem with major changes in fungal abundance, suggesting links with bee cold tolerance or overwintering biology. We find that A. bomboides hosts a conserved core microbiome that may provide key fitness advantages through larval development and diapause, which raises the question of how this microbiome is maintained and faithfully transmitted between generations. Our results suggest that focus on microbiomes of mature or active insect developmental stages may overlook stage-specific symbionts and microbial fitness contributions during host dormancy.


Subject(s)
Bacteria , Fungi , Symbiosis , Animals , Bees/microbiology , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Bacteria/growth & development , Fungi/physiology , Fungi/classification , Fungi/genetics , Fungi/isolation & purification , Fungi/growth & development , Larva/microbiology , Gastrointestinal Microbiome , Seasons , Host Microbial Interactions , Diapause/physiology
20.
Arch Microbiol ; 206(6): 253, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727738

ABSTRACT

Candida auris is an invasive fungal pathogen of high concern due to acquired drug tolerance against antifungals used in clinics. The prolonged persistence on biotic and abiotic surfaces can result in onset of hospital outbreaks causing serious health threat. An in depth understanding of pathology of C. auris is highly desirable for development of efficient therapeutics. Non-coding RNAs play crucial role in fungal pathology. However, the information about ncRNAs is scanty to be utilized. Herein our aim is to identify long noncoding RNAs with potent role in pathobiology of C. auris. Thereby, we analyzed the transcriptomics data of C. auris infection in blood for identification of potential lncRNAs with regulatory role in determining invasion, survival or drug tolerance under infection conditions. Interestingly, we found 275 lncRNAs, out of which 253 matched with lncRNAs reported in Candidamine, corroborating for our accurate data analysis pipeline. Nevertheless, we obtained 23 novel lncRNAs not reported earlier. Three lncRNAs were found to be under expressed throughout the course of infection, in the transcriptomics data. 16 of potent lncRNAs were found to be coexpressed with coding genes, emphasizing for their functional role. Noteworthy, these ncRNAs are expressed from intergenic regions of the genes associated with transporters, metabolism, cell wall biogenesis. This study recommends for possible association between lncRNA expression and C. auris pathogenesis.


Subject(s)
Candida auris , Candidiasis , Host Microbial Interactions , RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/isolation & purification , Gene Expression Profiling , Computer Simulation , Genome-Wide Association Study , Candida auris/genetics , Candida auris/pathogenicity , Candidiasis/blood , Candidiasis/microbiology , Sepsis/microbiology , Host Microbial Interactions/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...