Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 537
Filter
Add more filters

Publication year range
1.
Nature ; 584(7821): 398-402, 2020 08.
Article in English | MEDLINE | ID: mdl-32759999

ABSTRACT

Land use change-for example, the conversion of natural habitats to agricultural or urban ecosystems-is widely recognized to influence the risk and emergence of zoonotic disease in humans1,2. However, whether such changes in risk are underpinned by predictable ecological changes remains unclear. It has been suggested that habitat disturbance might cause predictable changes in the local diversity and taxonomic composition of potential reservoir hosts, owing to systematic, trait-mediated differences in species resilience to human pressures3,4. Here we analyse 6,801 ecological assemblages and 376 host species worldwide, controlling for research effort, and show that land use has global and systematic effects on local zoonotic host communities. Known wildlife hosts of human-shared pathogens and parasites overall comprise a greater proportion of local species richness (18-72% higher) and total abundance (21-144% higher) in sites under substantial human use (secondary, agricultural and urban ecosystems) compared with nearby undisturbed habitats. The magnitude of this effect varies taxonomically and is strongest for rodent, bat and passerine bird zoonotic host species, which may be one factor that underpins the global importance of these taxa as zoonotic reservoirs. We further show that mammal species that harbour more pathogens overall (either human-shared or non-human-shared) are more likely to occur in human-managed ecosystems, suggesting that these trends may be mediated by ecological or life-history traits that influence both host status and tolerance to human disturbance5,6. Our results suggest that global changes in the mode and the intensity of land use are creating expanding hazardous interfaces between people, livestock and wildlife reservoirs of zoonotic disease.


Subject(s)
Biodiversity , Host Specificity , Zoonoses/microbiology , Zoonoses/parasitology , Zoonoses/virology , Animals , Birds/microbiology , Birds/parasitology , Birds/virology , Humans , Mammals/microbiology , Mammals/parasitology , Mammals/virology , Species Specificity , Zoonoses/transmission
2.
Annu Rev Microbiol ; 74: 673-691, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32689912

ABSTRACT

Amyloids are implicated in many protein misfolding diseases. Amyloid folds, however, also display a range of functional roles particularly in the microbial world. The templating ability of these folds endows them with specific properties allowing their self-propagation and protein-to-protein transmission in vivo. This property, the prion principle, is exploited by specific signaling pathways that use transmission of the amyloid fold as a way to convey information from a receptor to an effector protein. I describe here amyloid signaling pathways involving fungal nucleotide binding and oligomerization domain (NOD)-like receptors that were found to control nonself recognition and programmed cell death processes. Studies on these fungal amyloid signaling motifs stem from the characterization of the fungal [Het-s] prion protein and have led to the identification in fungi but also in multicellular bacteria of several distinct families of signaling motifs, one of which is related to RHIM [receptor-interacting protein (RIP) homotypic interaction motif], an amyloid motif regulating mammalian necroptosis.


Subject(s)
Amyloid/metabolism , Bacteria/metabolism , Fungal Proteins/metabolism , Fungi/metabolism , Signal Transduction , Amyloid/chemistry , Animals , Bacteria/genetics , Fungal Proteins/genetics , Fungi/genetics , Mammals/microbiology , Models, Molecular , NLR Proteins/genetics , NLR Proteins/metabolism , Necroptosis , Prions/genetics , Prions/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
3.
Mol Biol Evol ; 40(7)2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37402639

ABSTRACT

Social networks can influence the ecology of gut bacteria, shaping the species composition of the gut microbiome in humans and other animals. Gut commensals evolve and can adapt at a rapid pace when colonizing healthy hosts. Here, we aimed at assessing the impact of host-to-host bacterial transmission on Escherichia coli evolution in the mammalian gut. Using an in vivo experimental evolution approach in mice, we found a transmission rate of 7% (±3% 2× standard error [2SE]) of E. coli cells per day between hosts inhabiting the same household. Consistent with the predictions of a simple population genetics model of mutation-selection-migration, the level of shared events resulting from within host evolution is greatly enhanced in cohoused mice, showing that hosts undergoing the same diet and habit are not only expected to have similar microbiome species compositions but also similar microbiome evolutionary dynamics. Furthermore, we estimated the rate of mutation accumulation of E. coli to be 3.0 × 10-3 (±0.8 × 10-3 2SE) mutations/genome/generation, irrespective of the social context of the regime. Our results reveal the impact of bacterial migration across hosts in shaping the adaptive evolution of new strains colonizing gut microbiomes.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Humans , Animals , Mice , Biological Evolution , Escherichia coli/genetics , Microbiota/genetics , Gastrointestinal Microbiome/genetics , Mutation , Mammals/microbiology , Bacteria
4.
Mol Biol Evol ; 40(7)2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37326290

ABSTRACT

How host-associated microbial communities evolve as their hosts diversify remains equivocal: how conserved is their composition? What was the composition of ancestral microbiota? Do microbial taxa covary in abundance over millions of years? Multivariate phylogenetic models of trait evolution are key to answering similar questions for complex host phenotypes, yet they are not directly applicable to relative abundances, which usually characterize microbiota. Here, we extend these models in this context, thereby providing a powerful approach for estimating phylosymbiosis (the extent to which closely related host species harbor similar microbiota), ancestral microbiota composition, and integration (evolutionary covariations in bacterial abundances). We apply our model to the gut microbiota of mammals and birds. We find significant phylosymbiosis that is not entirely explained by diet and geographic location, indicating that other evolutionary-conserved traits shape microbiota composition. We identify main shifts in microbiota composition during the evolution of the two groups and infer an ancestral mammalian microbiota consistent with an insectivorous diet. We also find remarkably consistent evolutionary covariations among bacterial orders in mammals and birds. Surprisingly, despite the substantial variability of present-day gut microbiota, some aspects of their composition are conserved over millions of years of host evolutionary history.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Phylogeny , Gastrointestinal Microbiome/genetics , Vertebrates/genetics , Microbiota/genetics , Mammals/genetics , Mammals/microbiology , Birds/genetics , Bacteria/genetics , RNA, Ribosomal, 16S/genetics
5.
Mol Ecol ; 33(1): e16862, 2024 Jan.
Article in English | MEDLINE | ID: mdl-36786039

ABSTRACT

Different host species associate with distinct gut microbes in mammals, a pattern sometimes referred to as phylosymbiosis. However, the processes shaping this host specificity are not well understood. One model proposes that barriers to microbial transmission promote specificity by limiting microbial dispersal between hosts. This model predicts that specificity levels measured across microbes is correlated to transmission mode (vertical vs. horizontal) and individual dispersal traits. Here, we leverage two large publicly available gut microbiota data sets (1490 samples from 195 host species) to test this prediction. We found that host specificity varies widely across bacteria (i.e., there are generalist and specialist bacteria) and depends on transmission mode and dispersal ability. Horizontally-like transmitted bacteria equipped with traits that facilitate switches between host (e.g., tolerance to oxygen) were found to be less specific (more generalist) than microbes without those traits, for example, vertically-like inherited bacteria that are intolerant to oxygen. Altogether, our findings are compatible with a model in which limited microbial dispersal abilities foster host specificity.


Subject(s)
Gastrointestinal Microbiome , Animals , Mammals/microbiology , Host Specificity , Bacteria/genetics , Oxygen
6.
Proc Biol Sci ; 290(2011): 20232223, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-37964521

ABSTRACT

The gut microbiome composition of terrestrial vertebrates is known to converge in response to common specialized dietary strategies, like leaf-eating (folivory) or ant- and termite-eating (myrmecophagy). To date, such convergence has been studied in mammals and birds, but has been neglected in amphibians. Here, we analysed 15 anuran species (frogs and toads) representing five Neotropical families and demonstrated the compositional convergence of the gut microbiomes of distantly related myrmecophagous species. Specifically, we found that the gut microbial communities of bufonids and microhylids, which have independently evolved myrmecophagy, were significantly more similar than expected based on their hosts' evolutionary divergence. Conversely, we found that gut microbiome composition was significantly associated with host evolutionary history in some cases. For instance, the microbiome composition of Xenohyla truncata, one of the few known amphibians that eat fruits, was not different from those of closely related tree frogs with an arthropod generalist diet. Bacterial taxa overrepresented in myrmecophagous species relative to other host families include Paludibacter, Treponema, and Rikenellaceae, suggesting diet-mediated selection and prey-to-predator transmission likely driving the observed compositional convergence. This study provides a basis for examining the roles of the gut microbiome in host tolerance and sequestration of toxic alkaloids from ants and termites.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Humans , Animals , Biological Evolution , Mammals/microbiology , Anura , RNA, Ribosomal, 16S
7.
BMC Microbiol ; 23(1): 87, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36997846

ABSTRACT

Mammalian gastrointestinal microbiomes are highly variable, both within individuals and across populations, with changes linked to time and ageing being widely reported. Discerning patterns of change in wild mammal populations can therefore prove challenging. We used high-throughput community sequencing methods to characterise the microbiome of wild field voles (Microtus agrestis) from faecal samples collected across 12 live-trapping field sessions, and then at cull. Changes in α- and ß-diversity were modelled over three timescales. Short-term differences (following 1-2 days captivity) were analysed between capture and cull, to ascertain the degree to which the microbiome can change following a rapid change in environment. Medium-term changes were measured between successive trapping sessions (12-16 days apart), and long-term changes between the first and final capture of an individual (from 24 to 129 days). The short period between capture and cull was characterised by a marked loss of species richness, while over medium and long-term in the field, richness slightly increased. Changes across both short and long timescales indicated shifts from a Firmicutes-dominant to a Bacteroidetes-dominant microbiome. Dramatic changes following captivity indicate that changes in microbiome diversity can be rapid, following a change of environment (food sources, temperature, lighting etc.). Medium- and long-term patterns of change indicate an accrual of gut bacteria associated with ageing, with these new bacteria being predominately represented by Bacteroidetes. While the patterns of change observed are unlikely to be universal to wild mammal populations, the potential for analogous shifts across timescales should be considered whenever studying wild animal microbiomes. This is especially true if studies involve animal captivity, as there are potential ramifications both for animal health, and the validity of the data itself as a reflection of a 'natural' state of an animal.


Subject(s)
Microbiota , Rodentia , Animals , Microbiota/genetics , Animals, Wild/microbiology , Bacteria/genetics , Mammals/microbiology , Bacteroidetes/genetics
8.
Annu Rev Microbiol ; 72: 399-422, 2018 09 08.
Article in English | MEDLINE | ID: mdl-29927706

ABSTRACT

Mammalian immune systems evolved within a diverse world dominated by microbes, making interactions between these two life-forms inevitable. Adaptive immunity protects against microbes through antigen-specific responses. In classical studies, these responses were investigated in the context of pathogenicity; however, we now know that they have significant effects on our resident microbes. In turn, microbes employ an arsenal of mechanisms to influence development and specificity of host immunity. Understanding these complex reactions will be necessary to develop microbiota-based strategies to prevent or treat disease. Here we review the literature detailing the cross talk between resident microbes with a focus on the specificity of host responses and the microbial molecules that influence them.


Subject(s)
Adaptive Immunity , Host-Pathogen Interactions , Immunity, Innate , Mammals/immunology , Mammals/microbiology , Microbiota/immunology , Animals , Humans
9.
Nature ; 548(7665): 82-86, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28770842

ABSTRACT

Anthrax is a globally important animal disease and zoonosis. Despite this, our current knowledge of anthrax ecology is largely limited to arid ecosystems, where outbreaks are most commonly reported. Here we show that the dynamics of an anthrax-causing agent, Bacillus cereus biovar anthracis, in a tropical rainforest have severe consequences for local wildlife communities. Using data and samples collected over three decades, we show that rainforest anthrax is a persistent and widespread cause of death for a broad range of mammalian hosts. We predict that this pathogen will accelerate the decline and possibly result in the extirpation of local chimpanzee (Pan troglodytes verus) populations. We present the epidemiology of a cryptic pathogen and show that its presence has important implications for conservation.


Subject(s)
Animal Diseases/mortality , Animals, Wild/microbiology , Anthrax/veterinary , Bacillus anthracis/pathogenicity , Mammals/microbiology , Rainforest , Tropical Climate , Africa South of the Sahara , Animal Diseases/microbiology , Animals , Anthrax/microbiology , Anthrax/mortality , Bacillus anthracis/isolation & purification , Diptera/microbiology , Extinction, Biological , Female , Male , Pan troglodytes/microbiology , Parks, Recreational , Phylogeny
10.
Proc Natl Acad Sci U S A ; 116(8): 3036-3041, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718421

ABSTRACT

The repeated evolution of dietary specialization represents a hallmark of mammalian ecology. To detect genomic changes that are associated with dietary adaptations, we performed a systematic screen for convergent gene losses associated with an obligate herbivorous or carnivorous diet in 31 placental mammals. For herbivores, our screen discovered the repeated loss of the triglyceride lipase inhibitor PNLIPRP1, suggesting enhanced triglyceride digestion efficiency. Furthermore, several herbivores lost the pancreatic exocytosis factor SYCN, providing an explanation for continuous pancreatic zymogen secretion in these species. For carnivores, we discovered the repeated loss of the hormone-receptor pair INSL5-RXFP4 that regulates appetite and glucose homeostasis, which likely relates to irregular feeding patterns and constant gluconeogenesis. Furthermore, reflecting the reduced need to metabolize plant-derived xenobiotics, several carnivores lost the xenobiotic receptors NR1I3 and NR1I2 Finally, the carnivore-associated loss of the gastrointestinal host defense gene NOX1 could be related to a reduced gut microbiome diversity. By revealing convergent gene losses associated with differences in dietary composition, feeding patterns, and gut microbiomes, our study contributes to understanding how similar dietary specializations evolved repeatedly in mammals.


Subject(s)
Carnivory/physiology , Gastrointestinal Tract/microbiology , Herbivory/genetics , Phylogeny , Animals , Diet , Female , Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/metabolism , Genome/genetics , Herbivory/physiology , Mammals/microbiology , Plants , Pregnancy , RNA, Ribosomal, 16S/genetics
11.
Proc Natl Acad Sci U S A ; 116(47): 23588-23593, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685619

ABSTRACT

A major challenge in biology is to understand how phylogeny, diet, and environment shape the mammalian gut microbiome. Yet most studies of nonhuman microbiomes have relied on relatively coarse dietary categorizations and have focused either on individual wild populations or on captive animals that are sheltered from environmental pressures, which may obscure the effects of dietary and environmental variation on microbiome composition in diverse natural communities. We analyzed plant and bacterial DNA in fecal samples from an assemblage of 33 sympatric large-herbivore species (27 native, 6 domesticated) in a semiarid East African savanna, which enabled high-resolution assessment of seasonal variation in both diet and microbiome composition. Phylogenetic relatedness strongly predicted microbiome composition (r = 0.91) and was weakly but significantly correlated with diet composition (r = 0.20). Dietary diversity did not significantly predict microbiome diversity across species or within any species except kudu; however, diet composition was significantly correlated with microbiome composition both across and within most species. We found a spectrum of seasonal sensitivity at the diet-microbiome nexus: Seasonal changes in diet composition explained 25% of seasonal variation in microbiome composition across species. Species' positions on (and deviations from) this spectrum were not obviously driven by phylogeny, body size, digestive strategy, or diet composition; however, domesticated species tended to exhibit greater diet-microbiome turnover than wildlife. Our results reveal marked differences in the influence of environment on the degree of diet-microbiome covariation in free-ranging African megafauna, and this variation is not well explained by canonical predictors of nutritional ecology.


Subject(s)
Animals, Wild/microbiology , Diet , Gastrointestinal Microbiome , Mammals/microbiology , Animals , Animals, Domestic/microbiology , Animals, Domestic/physiology , Animals, Wild/physiology , DNA, Bacterial/analysis , DNA, Plant/analysis , Feces/chemistry , Feces/microbiology , Herbivory , Kenya , Mammals/physiology , Models, Biological , Phylogeny , Plants, Edible , Ruminants/microbiology , Ruminants/physiology , Seasons , Species Specificity
12.
Infect Immun ; 89(10): e0030721, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34310888

ABSTRACT

Riboflavin is an essential micronutrient, but its transport and utilization have remained largely understudied among pathogenic spirochetes. Here, we show that Borrelia burgdorferi, the zoonotic spirochete that causes Lyme disease, is able to import riboflavin via products of its rfuABCD-like operon as well as synthesize flavin mononucleotide and flavin adenine dinucleotide despite lacking canonical genes for their synthesis. Additionally, a mutant deficient in the rfuABCD-like operon is resistant to the antimicrobial effect of roseoflavin, a natural riboflavin analog, and is attenuated in a murine model of Lyme borreliosis. Our combined results indicate not only that are riboflavin and the maintenance of flavin pools essential for B. burgdorferi growth but also that flavin utilization and its downstream products (e.g., flavoproteins) may play a more prominent role in B. burgdorferi pathogenesis than previously appreciated.


Subject(s)
Bacterial Proteins/genetics , Borrelia burgdorferi/drug effects , Borrelia burgdorferi/genetics , Lyme Disease/drug therapy , Lyme Disease/microbiology , Operon/genetics , Riboflavin/pharmacology , Animals , Female , Mammals/microbiology , Mice , Mice, Inbred C3H
13.
Mol Biol Evol ; 37(10): 3003-3022, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32467975

ABSTRACT

Dental calculus, the calcified form of the mammalian oral microbial plaque biofilm, is a rich source of oral microbiome, host, and dietary biomolecules and is well preserved in museum and archaeological specimens. Despite its wide presence in mammals, to date, dental calculus has primarily been used to study primate microbiome evolution. We establish dental calculus as a valuable tool for the study of nonhuman host microbiome evolution, by using shotgun metagenomics to characterize the taxonomic and functional composition of the oral microbiome in species as diverse as gorillas, bears, and reindeer. We detect oral pathogens in individuals with evidence of oral disease, assemble near-complete bacterial genomes from historical specimens, characterize antibiotic resistance genes, reconstruct components of the host diet, and recover host genetic profiles. Our work demonstrates that metagenomic analyses of dental calculus can be performed on a diverse range of mammalian species, which will allow the study of oral microbiome and pathogen evolution from a comparative perspective. As dental calculus is readily preserved through time, it can also facilitate the quantification of the impact of anthropogenic changes on wildlife and the environment.


Subject(s)
Dental Calculus/microbiology , Mammals/microbiology , Microbiota , Mouth/microbiology , Animals , Biological Evolution , Diet , Drug Resistance, Microbial/genetics , Gorilla gorilla , Metagenome , Reindeer , Ursidae
14.
Mol Biol Evol ; 37(2): 307-319, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31638147

ABSTRACT

Whether and how epigenetic mechanisms and the microbiome play a role in mammalian adaptation raised considerable attention and controversy, mainly because they have the potential to add new insights into the Modern Synthesis. Recent attempts to reconcile neo-Darwinism and neo-Lamarckism in a unified theory of molecular evolution give epigenetic mechanisms and microbiome a prominent role. However, supporting empirical data are still largely missing. Because experimental studies using extant animals can hardly be done over evolutionary timescales, we propose that advances in ancient DNA techniques provide a valid alternative. In this piece, we evaluate 1) the possible roles of epigenomes and microbiomes in animal adaptation, 2) advances in the retrieval of paleoepigenome and paleomicrobiome data using ancient DNA techniques, and 3) the plasticity of either and interactions between the epigenome and the microbiome, while emphasizing that it is essential to take both into account, as well as the underlying genetic factors that may confound the findings. We propose that advanced ancient DNA techniques should be applied to a wide range of past animals, so novel dynamics in animal evolution and adaption can be revealed.


Subject(s)
Adaptation, Physiological , DNA, Ancient/analysis , Epigenomics/methods , Mammals/genetics , Mammals/microbiology , Animals , Confounding Factors, Epidemiologic , DNA Methylation , Epigenesis, Genetic , Evolution, Molecular , Mammals/physiology , Microbiota , Paleontology
15.
Appl Environ Microbiol ; 87(11)2021 05 11.
Article in English | MEDLINE | ID: mdl-33741637

ABSTRACT

Borrelia persica, transmitted by the argasid tick Ornithodoros tholozani, causes human tick-borne relapsing fever in the Middle East and Central Asia. Infection is acquired often when visiting tick-infested caves and reported to be transmitted mainly transovarially between ticks, occasionally infecting humans. To study the epidemiology of this infection, ticks were trapped in 24 caves in 12 geographic zones covering all of Israel and identified morphologically. DNA was extracted from larvae, nymphs, and adult stages from each location and PCR followed by DNA sequencing was performed to identify Borrelia infection, tick species, and tick blood meal sources. We collected 51,472 argasid ticks from 16 of 24 caves surveyed. We analyzed 2,774 O. tholozani ticks, and 72 (2.6%) from nine caves were PCR positive for B. persica Infection rates in male, female, and nymphal ticks (4.4%, 3%, and 3.2%, respectively) were higher than in larva (P < 0.001), with only 3 (0.04%) positive larvae. Presence of blood meal was associated with B. persica infection in ticks (P = 0.003), and blood meals of golden jackals, red foxes, and Cairo spiny mouse were associated with infection (P ≤ 0.043). PCR survey of 402 wild mammals revealed B. persica infection with the highest rates in social voles (22%), red foxes (16%), golden jackals (8%), and Cairo spiny mice (3%). In conclusion, although transovarial tick transmission of B. persica occurs at low levels, ticks apparently acquire infection mainly from wildlife canid and rodents and may eventually transmit relapsing fever borreliosis to humans who enter their habitat.IMPORTANCEBorrelia persica is a spirochete that causes tick-borne relapsing fever in humans in an area that spans from India to the Mediterranean. Until now, it was thought that the soft tick vector of this infection, Ornithodoros tholozani, is also its main reservoir and it transmits B. persica mostly transovarially between tick generations. This study showed that tick infection with B. persica is associated with feeding blood from wild jackals, foxes, and rodents and that transovarial transmission is minimal. Since O. tholozani ticks are found in isolated caves and ruins, it is assumed that wild canids who migrate over long distances have a major role in the transmission of B. persica between remote tick populations, and it is then maintained locally also by rodents and eventually transferred to humans during tick bites. Prevention of human infection could be achieved by restricting entrance of canines and humans to habitats with O. tholozani populations.


Subject(s)
Bacterial Zoonoses/transmission , Borrelia/physiology , Disease Reservoirs/veterinary , Ornithodoros/physiology , Relapsing Fever/transmission , Animals , Animals, Wild/microbiology , Bacterial Zoonoses/microbiology , Birds/microbiology , Caves/parasitology , Diet , Disease Reservoirs/microbiology , Feeding Behavior , Female , Israel , Male , Mammals/microbiology , Nymph/growth & development , Nymph/microbiology , Nymph/physiology , Ornithodoros/growth & development , Ornithodoros/microbiology , Relapsing Fever/microbiology
16.
BMC Microbiol ; 21(1): 298, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34715771

ABSTRACT

BACKGROUND: Akkermansia muciniphila is a member of the human gut microbiota where it resides in the mucus layer and uses mucin as the sole carbon, nitrogen and energy source. A. muciniphila is the only representative of the Verrucomicrobia phylum in the human gut. However, A. muciniphila 16S rRNA gene sequences have also been found in the intestines of many vertebrates. RESULTS: We detected A. muciniphila-like bacteria in the intestines of animals belonging to 15 out of 16 mammalian orders. In addition, other species belonging to the Verrucomicrobia phylum were detected in fecal samples. We isolated 10 new A. muciniphila strains from the feces of chimpanzee, siamang, mouse, pig, reindeer, horse and elephant. The physiology and genome of these strains were highly similar in comparison to the type strain A. muciniphila MucT. Overall, the genomes of the new strains showed high average nucleotide identity (93.9 to 99.7%). In these genomes, we detected considerable conservation of at least 75 of the 78 mucin degradation genes that were previously detected in the genome of the type strain MucT. CONCLUSIONS: The low genomic divergence observed in the new strains may indicate that A. muciniphila favors mucosal colonization independent of the differences in hosts. In addition, the conserved mucus degradation capability points towards a similar beneficial role of the new strains in regulating host metabolic health.


Subject(s)
Genome, Bacterial/genetics , Mammals/microbiology , Akkermansia/classification , Akkermansia/genetics , Akkermansia/isolation & purification , Akkermansia/metabolism , Animals , Feces/microbiology , Gastrointestinal Tract/microbiology , Genetic Variation , Genomics , Humans , Mammals/classification , Mice , Mucins/metabolism , Phylogeny , RNA, Ribosomal, 16S/genetics , Verrucomicrobia/classification , Verrucomicrobia/genetics , Verrucomicrobia/isolation & purification
17.
BMC Microbiol ; 21(1): 180, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34126929

ABSTRACT

BACKGROUND: In the last few decades, considerable attention has been paid to entomopathogenic fungi as biocontrol agents, however little is known about their mode of action and safety. This study aimed to investigate the toxicity of Aspergillus flavus in insect Spodoptera litura by analyzing the effect of fungal extract on antioxidant and cellular immune defense. In antioxidant defense, the lipid peroxidation (Malondialdehyde content) and antioxidant enzymes activities (Catalase, Ascorbate peroxidase, Superoxide dismutase) were examined. In cellular immune defense, effect of A. flavus extract was analyzed on haemocytes using Scanning Electron Microscopy (SEM). Furthermore, mammalian toxicity was analyzed with respect to DNA damage induced in treated rat relative to control by comet assay using different tissues of rat (blood, liver, and kidney). RESULTS: Ethyl acetate extract of A. flavus was administrated to the larvae of S.litura using artificial diet method having concentration 1340.84 µg/ml (LC50 of fungus). The effect was observed using haemolymph of insect larvae for different time intervals (24, 48, 72 and 96). In particular, Malondialdehyde content and antioxidant enzymes activities were found to be significantly (p ≤ 0.05) increased in treated larvae as compared to control. A. flavus ethyl acetate extract also exhibit negative impact on haemocytes having major role in cellular immune defense. Various deformities were observed in different haemocytes like cytoplasmic leakage and surface abnormalities etc. Genotoxicity on rat was assessed using different tissues of rat (blood, liver, and kidney) by comet assay. Non-significant effect of A. flavus extract was found in all the tissues (blood, liver, and kidney). CONCLUSIONS: Overall the study provides important information regarding the oxidative stress causing potential and immunosuppressant nature of A. flavus against S. litura and its non toxicity to mammals (rat), mammals (rat), suggesting it an environment friendly pest management agent.


Subject(s)
Aspergillus flavus/physiology , Mammals/metabolism , Mammals/microbiology , Oxidative Stress , Spodoptera/microbiology , Animals , DNA Damage , Kidney/immunology , Kidney/metabolism , Kidney/microbiology , Larva/genetics , Larva/immunology , Larva/metabolism , Larva/microbiology , Liver/immunology , Liver/metabolism , Liver/microbiology , Male , Malondialdehyde/metabolism , Mammals/genetics , Mammals/immunology , Rats , Rats, Wistar , Spodoptera/genetics , Spodoptera/immunology , Spodoptera/metabolism
18.
Cell Microbiol ; 22(4): e13167, 2020 04.
Article in English | MEDLINE | ID: mdl-32185902

ABSTRACT

A fundamental question of eukaryotic cell biology is how membrane organelles are organised and interact with each other. Cell biologists address these questions by characterising the structural features of membrane compartments and the mechanisms that coordinate their exchange. To do so, they must rely on variety of cargo molecules and treatments that enable targeted perturbation, localisation, and labelling of specific compartments. In this context, bacterial toxins emerged in cell biology as paradigm shifting molecules that enabled scientists to not only study them from the side of bacterial infection but also from the side of the mammalian host. Their selectivity, potency, and versatility made them exquisite tools for uncovering much of our current understanding of membrane trafficking mechanisms. Here, we will follow the steps that lead toxins until their intracellular targets, highlighting how specific events helped us comprehend membrane trafficking and establish the fundamentals of various cellular organelles and processes. Bacterial toxins will continue to guide us in answering crucial questions in cellular biology while also acting as probes for new technologies and applications.


Subject(s)
Bacterial Toxins/metabolism , Cell Membrane/physiology , Host Microbial Interactions , Mammals/physiology , Animals , Cell Biology , Cell Movement , Mammals/microbiology , Protein Transport
19.
Parasite Immunol ; 43(5): e12816, 2021 05.
Article in English | MEDLINE | ID: mdl-33368329

ABSTRACT

The bacterial pathogen Borrelia burgdorferi is the causative agent of Lyme disease and is transmitted to humans through an Ixodes tick vector. B. burgdorferi is able to survive in both mammalian and tick hosts through careful modulation of its gene expression. This allows B. burgdorferi to adapt to the environmental and nutritional changes that occur when it is transmitted between the two hosts. Distinct interactions between the spirochete and its host occur at every step of the enzootic cycle and dictate the ability of the spirochete to survive until the next stage of the cycle. Studying the interface between B. burgdorferi, the Ixodes tick vector and the natural mammalian reservoirs has been made significantly more feasible through the complete genome sequences of the organisms and the advent of high throughput screening technologies. Ultimately, a thorough investigation of the interplay between the two domains (and two phyla within one domain) is necessary in order to completely understand how the pathogen is transmitted.


Subject(s)
Arachnid Vectors/microbiology , Borrelia burgdorferi/physiology , Host Microbial Interactions/physiology , Ixodes/microbiology , Lyme Disease/microbiology , Mammals/microbiology , Animals , Arachnid Vectors/immunology , Borrelia burgdorferi/genetics , Gene Expression , Humans , Ixodes/immunology , Lyme Disease/epidemiology , Lyme Disease/transmission , Mammals/blood , Mammals/parasitology , Microbiota , Nymph/microbiology , Salivary Glands/microbiology
20.
Proc Natl Acad Sci U S A ; 115(25): E5786-E5795, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29871947

ABSTRACT

Skin is the largest organ of the body and represents the primary physical barrier between mammals and their external environment, yet the factors that govern skin microbial community composition among mammals are poorly understood. The objective of this research was to generate a skin microbiota baseline for members of the class Mammalia, testing the effects of host species, geographic location, body region, and biological sex. Skin from the back, torso, and inner thighs of 177 nonhuman mammals was sampled, representing individuals from 38 species and 10 mammalian orders. Animals were sampled from farms, zoos, households, and the wild. The DNA extracts from all skin swabs were amplified by PCR and sequenced, targeting the V3-V4 regions of bacterial and archaeal 16S rRNA genes. Previously published skin microbiome data from 20 human participants, sampled and sequenced using an identical protocol to the nonhuman mammals, were included to make this a comprehensive analysis. Human skin microbial communities were distinct and significantly less diverse than all other sampled mammalian orders. The factor most strongly associated with microbial community data for all samples was whether the host was a human. Within nonhuman samples, host taxonomic order was the most significant factor influencing skin microbiota, followed by the geographic location of the habitat. By comparing the congruence between host phylogeny and microbial community dendrograms, we observed that Artiodactyla (even-toed ungulates) and Perissodactyla (odd-toed ungulates) had significant congruence, providing evidence of phylosymbiosis between skin microbial communities and their hosts.


Subject(s)
Bacteria/genetics , Mammals/microbiology , Microbiota/genetics , Skin/microbiology , Symbiosis/genetics , Animals , DNA/genetics , Humans , Phylogeny , RNA, Ribosomal, 16S/genetics
SELECTION OF CITATIONS
SEARCH DETAIL