Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 544
Filtrar
1.
Microbiol Spectr ; 12(10): e0086424, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39162518

RESUMEN

Mammals host a remarkable diversity and abundance of gut microbes. Biosynthetic gene clusters (BGCs) in microbial genomes encode biologically active chemical products and play an important role in microbe-host interactions. Traditionally, the exploration of gut microbial metabolic functions has relied on the pure culture method. However, given the limited amounts of microbes being cultivated, insights into the metabolism of gut microbes in mammals continued to be very limited. In this study, we adopted a computational pipeline for mining the metagenomic data (named taxonomy-guided identification of biosynthetic gene clusters, TaxiBGC) to identify experimentally verified BGCs in 373 metagenomes across 53 mammalian species in an unbiased manner. We demonstrated that polyketides (PKs) and nonribosomal peptides (NRPs) are representative of mammals, and the products derived from them were associated with cell-cell communication and resistance to inflammation. Large carnivores had the highest number of BGCs, followed by large herbivores and small mammals. We also observed that the large mammals had more common BGCs that aid in the biosynthesis of a variety of natural products. However, small mammals not only had fewer BGCs but were also unique to each species. Our results provide novel insights into the mining of metagenomic data sets to identify active BGCs and their products across mammals.IMPORTANCEThe gut microbes host numerous biosynthetic gene clusters (BGCs) that biosynthesize natural products and impact the host's physiology. Historically, our understanding of BGCs in mammalian gut microbes was largely based on studies on cultured isolates; however, only a small fraction of mammal-associated microbes have been investigated. The biochemical diversity of the mammalian gut microbiota is poorly understood. Metagenomic sequencing contains data from a vast number of organisms and provides information on the total gene content of communities. Unfortunately, the existing BGC prediction tools are designed for individual microbial genomes. Recently, a BGC prediction tool called the taxonomy-guided identification of biosynthetic gene clusters (TaxiBGC) that directly mine the metagenome was developed. To gain new insights into the microbial metabolism, we used TaxiBGC to predict BGCs from 373 metagenomes across 53 mammalian species representing seven orders. Our findings elucidate the functional activities of complex microbial communities in the gut.


Asunto(s)
Microbioma Gastrointestinal , Mamíferos , Metagenoma , Metagenómica , Familia de Multigenes , Animales , Microbioma Gastrointestinal/genética , Mamíferos/microbiología , Bacterias/genética , Bacterias/clasificación , Bacterias/metabolismo , Policétidos/metabolismo , Minería de Datos , Productos Biológicos/metabolismo , Femenino , Vías Biosintéticas/genética
2.
Sci Rep ; 14(1): 15508, 2024 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-38969657

RESUMEN

The gut microbiome of wild animals is subject to various environmental influences, including those associated with human-induced alterations to the environment. We investigated how the gut microbiota of a synurbic rodent species, the striped field mouse (Apodemus agrarius), change in cities of varying sizes, seeking the urban microbiota signature for this species. Fecal samples for analysis were collected from animals living in non-urbanized areas and green spaces of different-sized cities (Poland). Metagenomic 16S rRNA gene sequencing and further bioinformatics analyses were conducted. Significant differences in the composition of gut microbiomes among the studied populations were found. However, the observed changes were dependent on local habitat conditions, without strong evidence of a correlation with the size of the urbanized area. The results suggest that ecological detachment from a more natural, non-urban environment does not automatically lead to the development of an "urban microbiome" model in the studied rodent. The exposure to the natural environment in green spaces may serve as a catalyst for microbiome transformations, providing a previously underestimated contribution to the maintenance of native gut microbial communities in urban mammals.


Asunto(s)
Heces , Microbioma Gastrointestinal , ARN Ribosómico 16S , Animales , ARN Ribosómico 16S/genética , Heces/microbiología , Ciudades , Murinae/microbiología , Ecosistema , Mamíferos/microbiología , Bacterias/clasificación , Bacterias/genética
3.
Infect Dis Poverty ; 13(1): 54, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38982550

RESUMEN

BACKGROUND: Rickettsia and related diseases have been identified as significant global public health threats. This study involved comprehensive field and systematic investigations of various rickettsial organisms in Yunnan Province. METHODS: Between May 18, 2011 and November 23, 2020, field investigations were conducted across 42 counties in Yunnan Province, China, encompassing small mammals, livestock, and ticks. Preliminary screenings for Rickettsiales involved amplifying the 16S rRNA genes, along with additional genus- or species-specific genes, which were subsequently confirmed through sequencing results. Sequence comparisons were carried out using the Basic Local Alignment Search Tool (BLAST). Phylogenetic relationships were analyzed using the default parameters in the Molecular Evolutionary Genetics Analysis (MEGA) program. The chi-squared test was used to assess the diversities and component ratios of rickettsial agents across various parameters. RESULTS: A total of 7964 samples were collected from small mammals, livestock, and ticks through Yunnan Province and submitted for screening for rickettsial organisms. Sixteen rickettsial species from the genera Rickettsia, Anaplasma, Ehrlichia, Neoehrlichia, and Wolbachia were detected, with an overall prevalence of 14.72%. Among these, 11 species were identified as pathogens or potential pathogens to humans and livestock. Specifically, 10 rickettsial organisms were widely found in 42.11% (24 out of 57) of small mammal species. High prevalence was observed in Dremomys samples at 5.60%, in samples from regions with latitudes above 4000 m or alpine meadows, and in those obtained from Yuanmou County. Anaplasma phagocytophilum and Candidatus Neoehrlichia mikurensis were broadly infecting multiple genera of animal hosts. In contrast, the small mammal genera Neodon, Dremomys, Ochotona, Anourosorex, and Mus were carrying individually specific rickettsial agents, indicating host tropism. There were 13 rickettsial species detected in 57.14% (8 out of 14) of tick species, with the highest prevalence (37.07%) observed in the genus Rhipicephalus. Eight rickettsial species were identified in 2375 livestock samples. Notably, six new Rickettsiales variants/strains were discovered, and Candidatus Rickettsia longicornii was unambiguously identified. CONCLUSIONS: This large-scale survey provided further insight into the high genetic diversity and overall prevalence of emerging Rickettsiales within endemic hotspots in Yunnan Province. The potential threats posed by these emerging tick-borne Rickettsiales to public health warrant attention, underscoring the need for effective strategies to guide the prevention and control of emerging zoonotic diseases in China.


Asunto(s)
Variación Genética , Filogenia , Rickettsiales , Garrapatas , China/epidemiología , Animales , Prevalencia , Rickettsiales/genética , Rickettsiales/aislamiento & purificación , Rickettsiales/clasificación , Garrapatas/microbiología , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/análisis , Ganado/microbiología , Infecciones por Rickettsia/epidemiología , Infecciones por Rickettsia/microbiología , Infecciones por Rickettsia/veterinaria , Rickettsia/aislamiento & purificación , Rickettsia/genética , Rickettsia/clasificación , Mamíferos/microbiología , Humanos
4.
Vet Med Sci ; 10(4): e1546, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39016692

RESUMEN

BACKGROUND: This study focuses on the AMR profiles in E. coli isolated from captive mammals at EcoZoo San Martín, Baños de Agua Santa, Ecuador, highlighting the role of wildlife as reservoirs of resistant bacteria. AIMS: The aim of this research is to investigate the antimicrobial resistance profiles of E. coli strains isolated from various species of captive mammals, emphasizing the potential zoonotic risks and the necessity for integrated AMR management strategies. MATERIALS & METHODS: A total of 189 fecal samples were collected from 70 mammals across 27 species. These samples were screened for E. coli, resulting in 90 identified strains. The resistance profiles of these strains to 16 antibiotics, including 10 ß-lactams and 6 non-ß-lactams, were determined using the disk diffusion method. Additionally, the presence of Extended-Spectrum Beta-Lactamase (ESBL) genes and other resistance genes was analyzed using PCR. RESULTS: Significant resistance was observed, with 52.22% of isolates resistant to ampicillin, 42.22% to ceftriaxone and cefuroxime, and 27.78% identified as ESBL-producing E. coli. Multiresistance (resistance to more than three antibiotic groups) was found in 35.56% of isolates. Carnivorous and omnivorous animals, particularly those with prior antibiotic treatments, were more likely to harbor resistant strains. DISCUSSION: These findings underscore the role of captive mammals as indicators of environmental AMR. The high prevalence of resistant E. coli in these animals suggests that zoos could be significant reservoirs for the spread of antibiotic-resistant bacteria. The results align with other studies showing that diet and antibiotic treatment history influence resistance profiles. CONCLUSION: The study highlights the need for an integrated approach involving veterinary care, habitat management, and public awareness to prevent captive wildlife from becoming reservoirs of antibiotic-resistant bacteria. Improved waste management practices and responsible antibiotic use are crucial to mitigate the risks of AMR in zoo environments and reduce zoonotic threats.


Asunto(s)
Animales de Zoológico , Antibacterianos , Farmacorresistencia Bacteriana , Escherichia coli , Mamíferos , Animales , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/aislamiento & purificación , Ecuador/epidemiología , Mamíferos/microbiología , Antibacterianos/farmacología , Infecciones por Escherichia coli/veterinaria , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/epidemiología , Heces/microbiología
5.
Int J Antimicrob Agents ; 64(3): 107251, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38906487

RESUMEN

Antimicrobial resistance (AMR) has emerged as a critical global health challenge. However, the significance of AMR is not limited to humans and domestic animals but extends to wildlife and the environment. Based on the analysis of > 200 peer-reviewed papers, this review provides comprehensive and current insights into the detection of clinically significant antimicrobial resistant bacteria and resistance genes in wild mammals, birds and reptiles worldwide. The review also examines the overlooked roles of wildlife in AMR emergence and transmission. In wildlife, AMR is potentially driven by anthropogenic activity, agricultural and environmental factors, and natural evolution. This review highlights the significance of AMR surveillance in wildlife, identifies species and geographical foci and gaps, and demonstrates the value of multifaceted One Health strategies if further escalation of AMR globally is to be curtailed.


Asunto(s)
Animales Salvajes , Antibacterianos , Bacterias , Farmacorresistencia Bacteriana , Salud Única , Animales , Animales Salvajes/microbiología , Farmacorresistencia Bacteriana/genética , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/clasificación , Bacterias/aislamiento & purificación , Antibacterianos/farmacología , Aves/microbiología , Humanos , Mamíferos/microbiología , Reptiles/microbiología , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/veterinaria , Infecciones Bacterianas/tratamiento farmacológico , Salud Global
6.
Nat Commun ; 15(1): 5341, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937464

RESUMEN

Gut microbiomes of mammals carry a complex symbiotic assemblage of microorganisms. Feeding newborn infants milk from the mammary gland allows vertical transmission of the parental milk microbiome to the offspring's gut microbiome. This has benefits, but also has hazards for the host population. Using mathematical models, we demonstrate that biparental vertical transmission enables deleterious microbial elements to invade host populations. In contrast, uniparental vertical transmission acts as a sieve, preventing these invasions. Moreover, we show that deleterious symbionts generate selection on host modifier genes that keep uniparental transmission in place. Since microbial transmission occurs during birth in placental mammals, subsequent transmission of the milk microbiome needs to be maternal to avoid the spread of deleterious elements. This paper therefore argues that viviparity and the hazards from biparental transmission of the milk microbiome, together generate selection against male lactation in placental mammals.


Asunto(s)
Microbioma Gastrointestinal , Lactancia , Simbiosis , Animales , Femenino , Masculino , Microbioma Gastrointestinal/fisiología , Leche/microbiología , Embarazo , Mamíferos/microbiología , Herencia Materna
7.
FEMS Microbiol Ecol ; 100(7)2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38857884

RESUMEN

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.


Asunto(s)
Microbiota , Animales , Migración Animal , Biodiversidad , Aves/microbiología , Cambio Climático , Interacciones Microbiota-Huesped , Mamíferos/microbiología
8.
Acta Vet Hung ; 72(2): 71-79, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38842941

RESUMEN

The spread of antibiotic resistance is one of the biggest challenges of our time, making it difficult to treat bacterial diseases. Pasteurella multocida is a widespread facultative pathogenic bacterium, which causes a wide range of diseases in both mammals and birds. In the present study, antibiotic susceptibility of 155 P. multocida strains were tested using the broth microdilution method to obtain the minimum inhibitory concentration (MIC) values for 15 antibiotics. The most effective antibiotics against pasteurellosis were ceftiofur, tetracycline, doxycycline, florfenicol and tilmicosin. Of the strains, 12 proved to be multi-drug resistant (MDR). To combat antibiotic resistance, it is important to establish a pre-treatment antibiotic susceptibility profile. A well-chosen antibiotic would not only make the treatment more successful but may also slow down the spread of resistance and the evolution of MDR strains.


Asunto(s)
Antiinfecciosos , Farmacorresistencia Bacteriana , Pasteurella multocida , Pasteurella multocida/efectos de los fármacos , Pasteurella multocida/aislamiento & purificación , Antiinfecciosos/farmacología , Pruebas de Sensibilidad Microbiana , Infecciones por Pasteurella/microbiología , Farmacorresistencia Bacteriana Múltiple , Aves/microbiología , Mamíferos/microbiología , Animales , Bovinos
9.
Acta Trop ; 256: 107282, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38861832

RESUMEN

The hard tick clade (Ixodidae) currently comprises 762 species worldwide (266 Prostriata and 496 Metastriata). A quarter of hard ticks are found in the Neotropical region, and 42 species have been documented in Colombia. Ixodidae species are important vectors of pathogens such as bacteria, helminths, protozoa, and viruses. In tick-borne diseases, vertebrate hosts perform an important role in the transmission, maintenance, and spread of pathogens. Colombia ranks sixth among countries with the highest mammal biodiversity, with a total of 548 species, where some of these species may be involved in pathogen transmission cycles with ticks as vectors. This research evaluated the presence of two genera of bacteria (Borrelia and Rickettsia) and the protozoan (Babesia) in ticks and mammals in the Orinoquia region of Colombia, establishing interaction networks. The information comes from 734 mammals (655 wild and 79 domestic), belonging to 59 species. Tick infestation (n = 1,805) was found with 14.85 % (n = 109) of the examined mammals and corresponds to nine tick species confirmed morphologically and molecularly. To detect pathogens 272 ticks were collected while feeding on 96 mammals; samples from 93 mammals were analyzed. The presence of borreliae from the relapsing fever group (RFG) and the Lyme disease group (LDG) were detected. Rickettsia spp. was detected in ticks and mammals, while Babesia bigemina was only detected in ticks. This research is the first to address the prevalence of zoonotic pathogens in domestic and wild mammals infested with hard ticks in the Department of Arauca, Colombia. Considering that reporting cases of infections with Babesia, Borrelia, and Rickettsia in Colombia is not mandatory, their impact on public health cannot be estimated. This highlights the importance of continuously detecting, confirming, and identifying these and other important pathogens within the "One Health" framework, as they have a significant economic and medical-veterinary impact globally.


Asunto(s)
Babesia , Borrelia , Interacciones Huésped-Patógeno , Ixodidae , Mamíferos , Rickettsia , Animales , Colombia , Mamíferos/parasitología , Mamíferos/microbiología , Rickettsia/aislamiento & purificación , Rickettsia/genética , Ixodidae/microbiología , Ixodidae/parasitología , Babesia/aislamiento & purificación , Borrelia/aislamiento & purificación , Borrelia/patogenicidad , Infestaciones por Garrapatas/veterinaria , Infestaciones por Garrapatas/epidemiología , Infestaciones por Garrapatas/parasitología , Enfermedades por Picaduras de Garrapatas/microbiología , Enfermedades por Picaduras de Garrapatas/epidemiología , Enfermedades por Picaduras de Garrapatas/transmisión , Enfermedades por Picaduras de Garrapatas/parasitología
10.
Microbiome ; 12(1): 79, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38711157

RESUMEN

BACKGROUND: Disturbances alter the diversity and composition of microbial communities. Yet a generalized empirical assessment of microbiome responses to disturbance across different environments is needed to understand the factors driving microbiome recovery, and the role of the environment in driving these patterns. RESULTS: To this end, we combined null models with Bayesian generalized linear models to examine 86 time series of disturbed mammalian, aquatic, and soil microbiomes up to 50 days following disturbance. Overall, disturbances had the strongest effect on mammalian microbiomes, which lost taxa and later recovered their richness, but not their composition. In contrast, following disturbance, aquatic microbiomes tended away from their pre-disturbance composition over time. Surprisingly, across all environments, we found no evidence of increased compositional dispersion (i.e., variance) following disturbance, in contrast to the expectations of the Anna Karenina Principle. CONCLUSIONS: This is the first study to systematically compare secondary successional dynamics across disturbed microbiomes, using a consistent temporal scale and modeling approach. Our findings show that the recovery of microbiomes is environment-specific, and helps to reconcile existing, environment-specific research into a unified perspective. Video Abstract.


Asunto(s)
Bacterias , Teorema de Bayes , Microbiota , Microbiología del Suelo , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Mamíferos/microbiología , Biodiversidad , Microbiología del Agua
11.
Braz J Microbiol ; 55(2): 1941-1948, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38691328

RESUMEN

Leptospira spp. are bacteria responsible for leptospirosis, a zoonotic disease with considerable impacts on the economy, animal health, and public health. This disease has a global distribution and is particularly prevalent in Brazil. Both rural and urban environments are habitats for Leptospira spp., which are primarily transmitted through contact with the urine of infected animals. Consequently, domestic and wild species can harbor these prokaryotes and serve as infection sources for other hosts. In the context of wild animals, there is a dearth of molecular studies elucidating the roles of various animal and bacterial species in the epidemiology of leptospirosis. Therefore, this study aimed to evaluate the presence of Leptospira spp. DNA in different species of free-living and captive wild animals and to assess the phylogenetic relationships of the identified microorganisms in Rio Grande do Sul, Brazil. The samples were evaluated for the presence of the gene lipL32 by polymerase chain reaction (PCR) and sequencing of the amplified fragment after which phylogenetic analyzes were carried out. DNA from Leptospira spp. was extracted from kidney tissue from wild animals (Mammalia class). Pathogenic Leptospira spp. DNA was detected in 9.6% (11/114) of the samples, originating from nine species of wild animals, including the white-eared opossum (Didelphis albiventris), skunk (Conepatus chinga), geoffroy's cat (Leopardus geoffroyi), margay (Leopardus wiedii), pampas fox (Lycalopex gymnocercus), capybara (Hydrochoerus hydrochaeris), common marmoset (Callithrix jacchus), neotropical river otter (Lontra longicaudis), and european hare (Lepus europaeus). Phylogenetic analysis revealed the presence of Leptospira borgpetersenii and Leptospira interrogans in these animals. This research is the first study contributing to the epidemiology of leptospirosis by identifying L. borgpetersenii and L. interrogans in free-living and captive wild animals in Rio Grande do Sul, Brazil, potentially acting as bacterial reservoirs. Additionally, our findings can inform sanitary measures for controlling and preventing the disease, thereby safeguarding public health.


Asunto(s)
Animales Salvajes , Leptospira interrogans , Leptospira , Leptospirosis , Filogenia , Animales , Brasil/epidemiología , Leptospirosis/microbiología , Leptospirosis/veterinaria , Leptospirosis/epidemiología , Animales Salvajes/microbiología , Leptospira/genética , Leptospira/aislamiento & purificación , Leptospira/clasificación , Leptospira interrogans/genética , Leptospira interrogans/clasificación , Leptospira interrogans/aislamiento & purificación , Mamíferos/microbiología , ADN Bacteriano/genética
12.
Vet Microbiol ; 294: 110130, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38820727

RESUMEN

The transmission of antibiotic-resistant bacteria among wild animal species may hold significant epidemiological implications. However, this issue is seldom explored due to the perceived complexity of these systems, which discourages experimental investigation. To address this knowledge gap, we chose a configuration of birds and mammals coexisting in an urban green area as a research model: the rook Corvus frugilegus and the striped field mouse Apodemus agrarius. The indirect transmission of antimicrobial-resistant bacteria between these species is possible because rodents inhabiting rook colonies frequently come into contact with the birds' faeces and pellets. The study was conducted in two cities in eastern Poland (Central Europe) - Lublin and Chelm. Among 71 Escherichia (E.) coli isolates studied, 19.7% showed resistance to from one to six of the antibiotics tested, with much higher prevalence of antibiotic-resistant bacteria in the birds (32%) than in the rodents (7%). Whole genome sequencing was performed on 10 selected E. coli isolates representing similar resistance phenotypes. The following antimicrobial resistance genes were detected: blaTEM-1b, tet(A), tet(B), aph(6)-Id, aph(3'')-Ib, aadA1, aadA2, catA1, floR, cmlA, sul2, sul3, dfrA14, and dfrA2. Birds from the same city and also from both neighbouring cities shared E. coli bacteria with the same sequence types, whereas isolates detected in birds were not found to have been transferred to the mammalian population, despite close contact. This demonstrates that even intensive exposure to sources of these pathogens does not necessarily lead to effective transmission of antibiotic-resistant E. coli strains between birds and mammals. Further efforts should be dedicated to investigating actual transmission of antimicrobial-resistant bacteria in various ecological systems, including those that are crucial for public health, such as urban environments. This will facilitate the development of more accurate models for epidemiological threats and the formulation of well-balanced decisions regarding the coexistence of humans and urban wildlife.


Asunto(s)
Animales Salvajes , Antibacterianos , Ciudades , Escherichia coli , Animales , Polonia/epidemiología , Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/aislamiento & purificación , Animales Salvajes/microbiología , Heces/microbiología , Farmacorresistencia Bacteriana , Aves/microbiología , Mamíferos/microbiología , Infecciones por Escherichia coli/veterinaria , Infecciones por Escherichia coli/transmisión , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/epidemiología , Cuervos/microbiología , Pruebas de Sensibilidad Microbiana , Roedores/microbiología
13.
Microbiol Mol Biol Rev ; 88(2): e0020222, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38587383

RESUMEN

SUMMARYEvery human being is presumed to be infected by the fungus Pneumocystis jirovecii at least once in his or her lifetime. This fungus belongs to a large group of species that appear to exclusively infect mammals, with P. jirovecii being the only one known to cause disease in humans. The mystery of P. jirovecii origin and speciation is just beginning to unravel. Here, we provide a review of the major steps of P. jirovecii evolution. The Pneumocystis genus likely originated from soil or plant-associated organisms during the period of Cretaceous ~165 million years ago and successfully shifted to mammals. The transition coincided with a substantial loss of genes, many of which are related to the synthesis of nutrients that can be scavenged from hosts or cell wall components that could be targeted by the mammalian immune system. Following the transition, the Pneumocystis genus cospeciated with mammals. Each species specialized at infecting its own host. Host specialization is presumably built at least partially upon surface glycoproteins, whose protogene was acquired prior to the genus formation. P. jirovecii appeared at ~65 million years ago, overlapping with the emergence of the first primates. P. jirovecii and its sister species P. macacae, which infects macaques nowadays, may have had overlapping host ranges in the distant past. Clues from molecular clocks suggest that P. jirovecii did not cospeciate with humans. Molecular evidence suggests that Pneumocystis speciation involved chromosomal rearrangements and the mounting of genetic barriers that inhibit gene flow among species.


Asunto(s)
Filogenia , Pneumocystis carinii , Humanos , Animales , Pneumocystis carinii/genética , Pneumocystis carinii/clasificación , Pneumocystis carinii/patogenicidad , Infecciones por Pneumocystis/microbiología , Pneumocystis/genética , Pneumocystis/clasificación , Evolución Molecular , Especificidad del Huésped , Neumonía por Pneumocystis/microbiología , Genoma Fúngico/genética , Mamíferos/microbiología , Evolución Biológica
14.
mBio ; 15(3): e0334223, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38299854

RESUMEN

The mammalian mouth is colonized by complex microbial communities, adapted to specific niches, and in homeostasis with the host. Individual microbes interact metabolically and rely primarily on nutrients provided by the host, with which they have potentially co-evolved along the mammalian lineages. The oral environment is similar across mammals, but the diversity, specificity, and evolution of community structure in related or interacting mammals are little understood. Here, we compared the oral microbiomes of dogs with those of wild wolves and humans. In dogs, we found an increased microbial diversity relative to wolves, possibly related to the transition to omnivorous nutrition following domestication. This includes a larger diversity of Patescibacteria than previously reported in any other oral microbiota. The oral microbes are most distinct at bacterial species or strain levels, with few if any shared between humans and canids, while the close evolutionary relationship between wolves and dogs is reflected by numerous shared taxa. More taxa are shared at higher taxonomic levels including with humans, supporting their more ancestral common mammalian colonization followed by diversification. Phylogenies of selected oral bacterial lineages do not support stable human-dog microbial transfers but suggest diversification along mammalian lineages (apes and canids). Therefore, despite millennia of cohabitation and close interaction, the host and its native community controls and limits the assimilation of new microbes, even if closely related. Higher resolution metagenomic and microbial physiological studies, covering a larger mammalian diversity, should help understand how oral communities assemble, adapt, and interact with their hosts.IMPORTANCENumerous types of microbes colonize the mouth after birth and play important roles in maintaining oral health. When the microbiota-host homeostasis is perturbed, proliferation of some bacteria leads to diseases such as caries and periodontitis. Unlike the gut microbiome, the diversity of oral microbes across the mammalian evolutionary space is not understood. Our study compared the oral microbiomes of wild wolves, dogs, and apes (humans, chimpanzees, and bonobos), with the aim of identifying if microbes have been potentially exchanged between humans and dogs as a result of domestication and cohabitation. We found little if any evidence for such exchanges. The significance of our research is in finding that the oral microbiota and/or the host limit the acquisition of exogenous microbes, which is important in the context of natural exclusion of potential novel pathogens. We provide a framework for expanded higher-resolution studies across domestic and wild animals to understand resistance/resilience.


Asunto(s)
Microbioma Gastrointestinal , Hominidae , Microbiota , Lobos , Humanos , Animales , Perros , Mamíferos/microbiología , Bacterias
15.
Mol Ecol ; 33(1): e16862, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36786039

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Animales , Mamíferos/microbiología , Especificidad del Huésped , Bacterias/genética , Oxígeno
16.
Gut Microbes ; 16(1): 2296603, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38149632

RESUMEN

The human gut microbiota constitutes a vast and complex community of microorganisms. The myriad of microorganisms present in the intestinal tract exhibits highly intricate interactions, which play a crucial role in maintaining the stability and balance of the gut microbial ecosystem. These interactions, in turn, influence the overall health of the host. The mammalian gut microbes have evolved a wide range of mechanisms to suppress or even eliminate their competitors for nutrients and space. Simultaneously, extensive cooperative interactions exist among different microbes to optimize resource utilization and enhance their own fitness. This review will focus on the competitive mechanisms among members of the gut microorganisms and discuss key modes of actions, including bacterial secretion systems, bacteriocins, membrane vesicles (MVs) etc. Additionally, we will summarize the current knowledge of the often-overlooked positive interactions within the gut microbiota, and showcase representative machineries. This information will serve as a reference for better understanding the complex interactions occurring within the mammalian gut environment. Understanding the interaction dynamics of competition and cooperation within the gut microbiota is crucial to unraveling the ecology of the mammalian gut microbial communities. Targeted interventions aimed at modulating these interactions may offer potential therapeutic strategies for disease conditions.


Asunto(s)
Bacteriocinas , Microbioma Gastrointestinal , Microbiota , Animales , Humanos , Interacciones Microbianas , Mamíferos/microbiología
17.
Proc Biol Sci ; 290(2011): 20232223, 2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-37964521

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Animales , Evolución Biológica , Mamíferos/microbiología , Anuros , ARN Ribosómico 16S
18.
mSystems ; 8(5): e0038823, 2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37650612

RESUMEN

IMPORTANCE: Myrmecophagous mammals are specialized in the consumption of ants and/or termites. They do not share a direct common ancestor and evolved convergently in five distinct placental orders raising questions about the underlying adaptive mechanisms involved and the relative contribution of natural selection and phylogenetic constraints. Understanding how these species digest their prey can help answer these questions. More specifically, the role of their gut microbial symbionts in the digestion of the insect chitinous exoskeleton has not been investigated in all myrmecophagous orders. We generated 29 new gut metagenomes from nine myrmecophagous species to reconstruct more than 300 bacterial genomes in which we identified chitin-degrading enzymes. Studying the distribution of these chitinolytic bacteria among hosts revealed both shared and specific bacteria between ant-eating species. Overall, our results highlight the potential role of gut symbionts in the convergent dietary adaptation of myrmecophagous mammals and the evolutionary mechanisms shaping their gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Embarazo , Animales , Femenino , Microbioma Gastrointestinal/genética , Filogenia , Quitina , Placenta , Mamíferos/microbiología , Digestión
19.
Mol Biol Evol ; 40(7)2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37402639

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Animales , Ratones , Evolución Biológica , Escherichia coli/genética , Microbiota/genética , Microbioma Gastrointestinal/genética , Mutación , Mamíferos/microbiología , Bacterias
20.
Mol Biol Evol ; 40(7)2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37326290

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Animales , Filogenia , Microbioma Gastrointestinal/genética , Vertebrados/genética , Microbiota/genética , Mamíferos/genética , Mamíferos/microbiología , Aves/genética , Bacterias/genética , ARN Ribosómico 16S/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA