Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 544
Filtrar
1.
Microbiome ; 11(1): 249, 2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37953319

RESUMO

BACKGROUND: Reports regarding the presence of bacteria in the fetal environment remain limited and controversial. Recently, extracellular vesicles secreted by the human gut microbiota have emerged as a novel mechanism for host-microbiota interaction. We aimed to investigate the presence of bacterial extracellular vesicles in the fetal environment during healthy pregnancies and determine whether extracellular vesicles derived from the gut microbiota can cross biological barriers to reach the fetus. RESULTS: Bacterial extracellular vesicles were detectable in the amniotic fluid of healthy pregnant women, exhibiting similarities to extracellular vesicles found in the maternal gut microbiota. In pregnant mice, extracellular vesicles derived from human maternal gut microbiota were found to reach the intra-amniotic space. CONCLUSIONS: Our findings reveal maternal microbiota-derived extracellular vesicles as an interaction mechanism between the maternal microbiota and fetus, potentially playing a pivotal role in priming the prenatal immune system for gut colonization after birth. Video Abstract.


Assuntos
Vesículas Extracelulares , Microbioma Gastrointestinal , Microbiota , Gravidez , Feminino , Humanos , Camundongos , Animais , Feto/microbiologia , Líquido Amniótico/microbiologia , Bactérias
2.
Nature ; 613(7945): 639-649, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36697862

RESUMO

Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.


Assuntos
Biomassa , Contaminação por DNA , Feto , Microbiota , Animais , Feminino , Humanos , Gravidez , Líquido Amniótico/imunologia , Líquido Amniótico/microbiologia , Mamíferos , Microbiota/genética , Placenta/imunologia , Placenta/microbiologia , Feto/imunologia , Feto/microbiologia , Reprodutibilidade dos Testes
3.
Proc Natl Acad Sci U S A ; 119(49): e2212548119, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36442114

RESUMO

Microbial exposure during development can elicit long-lasting effects on the health of an individual. However, how microbial exposure in early life leads to permanent changes in the immune system is unknown. Here, we show that the microbial environment alters the set point for immune susceptibility by altering the developmental architecture of the CD8+ T cell compartment. In particular, early microbial exposure results in the preferential expansion of highly responsive fetal-derived CD8+ T cells that persist into adulthood and provide the host with enhanced immune protection against intracellular pathogens. Interestingly, microbial education of fetal-derived CD8+ T cells occurs during thymic development rather than in the periphery and involves the acquisition of a more effector-like epigenetic program. Collectively, our results provide a conceptual framework for understanding how microbial colonization in early life leads to lifelong changes in the immune system.


Assuntos
Linfócitos T CD8-Positivos , Feto , Imunidade , Diferenciação Celular , Escolaridade , Epigenômica , Feto/imunologia , Feto/microbiologia
4.
Braz J Microbiol ; 53(4): 2251-2262, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36303022

RESUMO

Opportunistic bacteria and fungi are commonly reported causes of bovine abortion in a small percentage of fetal losses of infectious etiology in cattle. The objective of this study was to characterize the pathological and etiological findings in fetuses aborted due to secondary bacterial and fungal infections submitted for postmortem examination between 2004 and 2019 in the State of Rio Grande do Sul, Brazil. Nineteen cases of bacterial etiology and five cases of fungal etiology were assessed. In cases of bacterial etiology, gross changes were uncommon and two different microscopic patterns were observed: (1) primary bronchopneumonia with occasional dissemination in cases of Staphylococcus sp., Streptococcus sp., and Mannheimia haemolytica infections; and (2) systemic disease with sepsis in cases of Escherichia coli and Listeria sp. infections. Aspergillus sp. was the main fungal agent identified, and cases of mycotic abortion were characterized by placentitis, dermatitis, and pneumonia. Fetal membranes were available for examination in less than half of the submissions (11/24), and placental lesions were observed in all cases. This study reaffirms the importance of postmortem examinations in the determination of causes of fetal loss in cattle and highlights pathological findings commonly observed in fetuses aborted due to sporadic bacterial and fungal agents.


Assuntos
Infecções Bacterianas , Doenças dos Bovinos , Micoses , Bovinos , Gravidez , Animais , Feminino , Humanos , Aborto Animal/microbiologia , Placenta/microbiologia , Placenta/patologia , Micoses/veterinária , Feto/microbiologia , Feto/patologia , Bactérias/genética , Doenças dos Bovinos/microbiologia , Infecções Bacterianas/veterinária
5.
PLoS One ; 17(1): e0262229, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35061805

RESUMO

Borrelia burgdorferi and Borrelia miyamotoi are tick-vectored zoonotic pathogens maintained in wildlife species. Tick populations are establishing in new areas globally in response to climate change and other factors. New Brunswick is a Canadian maritime province at the advancing front of tick population establishment and has seen increasing numbers of ticks carrying B. burgdorferi, and more recently B. miyamotoi. Further, it is part of a region of Atlantic Canada with wildlife species composition differing from much of continental North America and little information exists as to the presence and frequency of infection of Borrelia spp. in wildlife in this region. We used a citizen science approach to collect a wide range of animals including migratory birds, medium-sized mammals, and small mammals. In total we tested 339 animals representing 20 species for the presence of B. burgdorferi and B. miyamotoi. We have developed new nested PCR primers and a protocol with excellent specificity for detecting both of these Borrelia species, both single and double infections, in tissues and organs of various wildlife species. The positive animals were primarily small non-migratory mammals, approximately twice as many were infected with B. burgdorferi than B. miyamotoi and one animal was found infected with both. In addition to established reservoir species, the jumping mouse (Napaeozapus insignis) was found frequently infected; this species had the highest infection prevalence for both B. burgdorferi and B. miyamotoi and has not previously been identified as an important carrier for either Borrelia species. Comprehensive testing of tissues found that all instances of B. burgdorferi infection were limited to one tissue within the host, whereas two of the five B. miyamotoi infections were diffuse and found in multiple systems. In the one coinfected specimen, two fetuses were also recovered and found infected with B. miyamotoi. This presumptive transplacental transmission suggests that vertical transmission in mammals is possible. This finding implies that B. miyamotoi could rapidly spread into wildlife populations, as well as having potential human health implications.


Assuntos
Animais Selvagens/microbiologia , Borrelia burgdorferi/isolamento & purificação , Borrelia/isolamento & purificação , Animais , Aves/microbiologia , Borrelia/classificação , Borrelia/genética , Borrelia burgdorferi/classificação , Borrelia burgdorferi/genética , Canadá/epidemiologia , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Vetores de Doenças , Feto/microbiologia , Doença de Lyme/diagnóstico , Doença de Lyme/epidemiologia , Doença de Lyme/veterinária , Camundongos , Filogenia , Reação em Cadeia da Polimerase
6.
Gut Microbes ; 14(1): 2005751, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34923897

RESUMO

There is an ongoing controversy around the existence of a prenatal, fetal microbiome in humans, livestock, and other animals. The 'in utero microbial colonization' hypothesis challenges the clinical paradigm of the 'sterile womb' but has been criticized for its reliance on DNA-based evidence to detect microbiomes and the failure to conciliate the routine experimental derivation of germ-free animals from surgically resected embryos with a thriving fetal microbiome. In order to avoid the propagation of misinformation in the scientific literature, a critical assessment and careful review of newly published studies, particularly those that challenge the convincing current clinical dogma of the sterile womb, is of critical importance.We read with interest a recent publication that postulated the presence of a fetal microbiome in sheep, but questioned the plausibility of the reported findings and their meaningfulness to prove "microbial colonisation of the fetal gut […] in utero". We reanalyzed the published metagenomic and metatranscriptomic sequence data from the original publication and identified evidence for different types of contamination that affected all samples alike and could explain the reported findings without requiring the existence of a fetal microbiome.Our reanalysis challenges the reported findings as supportive of a prenatal fetal lamb microbiome. The shortcomings of the original analysis and data interpretation highlight common problems of low-biomass microbiome projects. We propose genomic independence of separate biological samples, i.e. distinctive profiles at the microbial strain level, as a potential new microbiome marker to increase confidence in metagenomics analyses of controversial low-biomass microbiomes.


Assuntos
Bactérias/isolamento & purificação , Feto/microbiologia , Microbioma Gastrointestinal , Ovinos/microbiologia , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Contaminação por DNA , Feminino , Gravidez
7.
Cell ; 184(13): 3394-3409.e20, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34077752

RESUMO

The human fetal immune system begins to develop early during gestation; however, factors responsible for fetal immune-priming remain elusive. We explored potential exposure to microbial agents in utero and their contribution toward activation of memory T cells in fetal tissues. We profiled microbes across fetal organs using 16S rRNA gene sequencing and detected low but consistent microbial signal in fetal gut, skin, placenta, and lungs in the 2nd trimester of gestation. We identified several live bacterial strains including Staphylococcus and Lactobacillus in fetal tissues, which induced in vitro activation of memory T cells in fetal mesenteric lymph node, supporting the role of microbial exposure in fetal immune-priming. Finally, using SEM and RNA-ISH, we visualized discrete localization of bacteria-like structures and eubacterial-RNA within 14th weeks fetal gut lumen. These findings indicate selective presence of live microbes in fetal organs during the 2nd trimester of gestation and have broader implications toward the establishment of immune competency and priming before birth.


Assuntos
Bactérias/metabolismo , Desenvolvimento Embrionário , Feto/citologia , Feto/microbiologia , Leucócitos/citologia , Adulto , Bactérias/genética , Bactérias/ultraestrutura , Proliferação de Células , Células Dendríticas/metabolismo , Feminino , Feto/ultraestrutura , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/ultraestrutura , Humanos , Memória Imunológica , Ativação Linfocitária/imunologia , Viabilidade Microbiana , Gravidez , Segundo Trimestre da Gravidez , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Reprodutibilidade dos Testes , Linfócitos T/citologia
8.
Nat Microbiol ; 6(7): 865-873, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33972766

RESUMO

Microbial colonization of the human intestine impacts host metabolism and immunity; however, exactly when colonization occurs is unclear. Although many studies have reported bacterial DNA in first-pass meconium samples, these samples are typically collected hours to days after birth. Here, we investigated whether bacteria could be detected in meconium before birth. Fetal meconium (n = 20) was collected by rectal swab during elective breech caesarean deliveries without labour and before antibiotics and compared to technical and procedural controls (n = 5), first-pass meconium (neonatal meconium; n = 14) and infant stool (n = 25). Unlike first-pass meconium, no microbial signal distinct from negative controls was detected in fetal meconium by 16S ribosomal RNA gene sequencing. Additionally, positive aerobic (n = 10 of 20) and anaerobic (n = 12 of 20) clinical cultures of fetal meconium (13 of 20 samples positive in at least one culture) were identified as likely skin contaminants, most frequently Staphylococcus epidermidis, and not detected by sequencing in most samples (same genera detected by culture and sequencing in 2 of 13 samples with positive culture). We conclude that fetal gut colonization of healthy term infants does not occur before birth and that microbial profiles of neonatal meconium reflect populations acquired during and after birth.


Assuntos
Feto/microbiologia , Mecônio/microbiologia , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Cesárea , Fezes/microbiologia , Feminino , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Masculino , Microbiota , Gravidez , Reto/microbiologia
9.
Gut ; 70(5): 853-864, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33589511

RESUMO

OBJECTIVE: Microbial exposure is critical to neonatal and infant development, growth and immunity. However, whether a microbiome is present in the fetal gut prior to birth remains debated. In this study, lambs delivered by aseptic hysterectomy at full term were used as an animal model to investigate the presence of a microbiome in the prenatal gut using a multiomics approach. DESIGN: Lambs were euthanised immediately after aseptic caesarean section and their cecal content and umbilical cord blood samples were aseptically acquired. Cecal content samples were assessed using metagenomic and metatranscriptomic sequencing to characterise any existing microbiome. Both sample types were analysed using metabolomics in order to detect microbial metabolites. RESULTS: We detected a low-diversity and low-biomass microbiome in the prenatal fetal gut, which was mainly composed of bacteria belonging to the phyla Proteobacteria, Actinobacteria and Firmicutes. Escherichia coli was the most abundant species in the prenatal fetal gut. We also detected multiple microbial metabolites including short chain fatty acids, deoxynojirimycin, mitomycin and tobramycin, further indicating the presence of metabolically active microbiota. Additionally, bacteriophage phiX174 and Orf virus, as well as antibiotic resistance genes, were detected in the fetal gut, suggesting that bacteriophage, viruses and bacteria carrying antibiotic resistance genes can be transmitted from the mother to the fetus during the gestation period. CONCLUSIONS: This study provides strong evidence that the prenatal gut harbours a microbiome and that microbial colonisation of the fetal gut commences in utero.


Assuntos
Feto/metabolismo , Feto/microbiologia , Microbioma Gastrointestinal/genética , Ovinos/genética , Ovinos/microbiologia , Animais , Feminino , Perfilação da Expressão Gênica , Metabolômica , Metagenômica , Modelos Animais , Gravidez
11.
Microbiome ; 9(1): 5, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436093

RESUMO

Within the last 6 years, a research field has emerged that focuses on the characterization of microbial communities in the prenatal intrauterine environment of humans and their putative role in human health. However, there is considerable controversy around the existence of such microbial populations. The often contentious debate is primarily focused on technical aspects of the research, such as difficulties to assure aseptic sampling and to differentiate legitimate signals in the data from contamination. Although such discussions are clearly important, we feel that the problems with the prenatal microbiome field go deeper. In this commentary, we apply a philosophical framework to evaluate the foundations, experimental approaches, and interpretations used by scientists on both sides of the debate. We argue that the evidence for a "sterile womb" is based on a scientific approach that aligns well with important principles of the philosophy of science as genuine tests of the hypothesis and multiple angles of explanatory considerations were applied. In contrast, research in support of the "in utero colonization hypothesis" is solely based on descriptive verifications that do not provide explanatory insight, which weakens the evidence for a prenatal intrauterine microbiome. We propose that a reflection on philosophical principles can inform not only the debate on the prenatal intrauterine microbiome but also other disciplines that attempt to study low-biomass microbial communities.


Assuntos
Feto/microbiologia , Microbiota , Filosofia , Útero/microbiologia , Feminino , Humanos , Modelos Biológicos , Gravidez , Reprodutibilidade dos Testes
12.
Microbiome ; 9(1): 8, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436098

RESUMO

For more than a century, the prenatal environment was considered sterile. Over the last few years, findings obtained with next-generation sequencing approaches from samples of the placenta, the amniotic fluid, meconium, and even fetal tissues have challenged the dogma of a sterile womb, and additional reports have emerged that used culture, microscopy, and quantitative PCR to support the presence of a low-biomass microbial community at prenatal sites. Given the substantial implications of prenatal exposure to microbes for the development and health of the host, the findings have gathered substantial interest from academics, high impact journals, the public press, and funding agencies. However, an increasing number of studies have challenged the prenatal microbiome identifying contamination as a major issue, and scientists that remained skeptical have pointed to inconsistencies with in utero colonization, the impact of c-sections on early microbiome assembly, and the ability to generate germ-free mammals. A lively academic controversy has emerged on the existence of the wider importance of prenatal microbial communities. Microbiome has asked experts to discuss these issues and provide their thoughts on the implications. To allow for a broader perspective of this discussion, we have specifically selected scientists, who have a long-standing expertise in microbiome sciences but who have not directly been involved in the debate so far.


Assuntos
Dissidências e Disputas , Feto/microbiologia , Microbiota/fisiologia , Modelos Biológicos , Líquido Amniótico/microbiologia , Animais , Feminino , Vida Livre de Germes , Humanos , Recém-Nascido , Mecônio/microbiologia , Placenta/microbiologia , Gravidez , Útero/microbiologia
13.
Microbiome ; 9(1): 7, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436100

RESUMO

The human microbiome refers to the genetic composition of microorganisms in a particular location in the human body. Emerging evidence over the past many years suggests that the microbiome constitute drivers of human fate almost at par with our genome and epigenome. It is now well accepted after decades of disbelief that a broad understanding of human development, health, physiology, and disease requires understanding of the microbiome along with the genome and epigenome. We are learning daily of the interdependent relationships between microbiome/microbiota and immune responses, mood, cancer progression, response to therapies, aging, obesity, antibiotic usage, and overusage and much more. The next frontier in microbiome field is understanding when does this influence begin? Does the human microbiome initiate at the time of birth or are developing human fetuses already primed with microbes and their products in utero. In this commentary, we reflect on evidence gathered thus far on this question and identify the unknown common truths. We present a way forward to continue understanding our microbial colleagues and our interwoven fates.


Assuntos
Consenso , Feto/microbiologia , Microbiota/fisiologia , Útero/microbiologia , Feminino , Humanos , Incerteza
14.
Sci Rep ; 11(1): 708, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436911

RESUMO

The fetus develops in a privileged environment, as the placenta serves as both a gateway for nutrients and a barrier for pathogen transfer to the fetus. Regardless, recent evidence suggests the presence of bacterial DNA in both placenta and fetus, and we have reported that DNA and protein from small numbers of bacteria gain access to the fetus from the maternal bloodstream. Other routes of environmental bacterial transfer from the mother to fetus remain unknown, as well as the physiological relevance of their presence. In these experiments, we examine multiple routes by which bacterial cellular components can enter the fetus and the fetal response to influx of bacterial DNA and protein. We inoculated maternal sheep with genetically-labeled S. aureus (Staphylococcus aureus) using three routes: intravenously, orally, and intra-vaginally. The inoculum did not produce sepsis or fever in the ewes, therefore mimicking incidental exposure to bacteria during pregnancy. 3-5 days post inoculation, we assessed the presence of bacterial components in the fetal tissues and analyzed fetal brain tissue to identify any alterations in gene expression. Our results demonstrate that components of bacteria that were introduced into the maternal mouth were detected in the fetal brain and that they stimulated changes in gene expression. We conclude that an oral route of transmission is relevant for transfer of bacterial cellular components to the fetus.


Assuntos
Encéfalo/microbiologia , DNA Bacteriano/análise , Feto/microbiologia , Troca Materno-Fetal , Placenta/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/isolamento & purificação , Administração Oral , Animais , Encéfalo/metabolismo , DNA Bacteriano/genética , Feminino , Feto/metabolismo , Regulação Bacteriana da Expressão Gênica , Transmissão Vertical de Doenças Infecciosas/estatística & dados numéricos , Placenta/metabolismo , Gravidez , RNA-Seq , Ovinos , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/metabolismo
15.
Acta Diabetol ; 58(3): 249-265, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32712802

RESUMO

Early-life healthy gut microbiota has a profound implication on shaping the mucosal immune system as well as maintaining healthy status later in life, especially at the prenatal or neonatal stages, while intestinal dysbiosis in early life is associated with several autoimmune diseases, including type 1 diabetes (T1D). Since the gut microbiome is potentially modifiable, optimizing the intestinal bacterial composition in early life may be a novel option for T1D prevention. In this review, we will review current data depicting the crucial role of early-life intestinal microbiome in the development of T1D and discuss the possible mechanisms whereby early-life intestinal microbiome influences the T1D progression. We also summarize recent findings on environmental factors affecting gut microbiota colonization and interventions that may successfully alter microbial composition to discuss potential means of preventing T1D progression in at-risk children.


Assuntos
Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 1/microbiologia , Feto/microbiologia , Microbioma Gastrointestinal/fisiologia , Recém-Nascido , Animais , Doenças Autoimunes/etiologia , Doenças Autoimunes/patologia , Criança , Diabetes Mellitus Tipo 1/imunologia , Disbiose/complicações , Disbiose/imunologia , Feminino , Feto/imunologia , Humanos , Recém-Nascido/imunologia , Masculino , Gravidez , Fatores de Risco
16.
Vet Q ; 41(1): 36-49, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33349157

RESUMO

BACKGROUND: Brucellosis of goats is caused by Brucella melitensis. It is a re-emerging zoonotic disease in many countries due to transmission from domestic animals and wildlife such as ibex, deer and wild buffaloes. OBJECTIVE: To describe the pathological changes, identification and distribution of B. melitensis in foetuses of experimentally infected does. METHODS: Twelve female goats of approximately 90 days pregnant were divided into 4 groups. Group 1 was exposed intra-conjunctival to 100 µL of sterile PBS while goats of Groups 2, 3 and 4 were similarly exposed to 100 µL of an inoculum containing 109 CFU/mL of live B. melitensis. Goats of these groups were killed at 15, 30 and 60 days post-inoculation, respectively. Foetal fluid and tissues were collected for bacterial identification (using direct bacterial culture, PCR and immuno-peroxidase staining) and histopathological examination. RESULTS: Bilateral intra-conjunctival exposure of pregnant does resulted in in-utero infection of the foetuses. All full-term foetuses of group 4 were either aborted or stillborn, showing petechiations of the skin or absence of hair coat with subcutaneous oedema. The internal organs showed most severe lesions. Immune-peroxidase staining revealed antigen distribution in all organs that became most extensive in group 4. Brucella melitensis was successfully isolated from the stomach content, foetal fluid and various other organs. CONCLUSION: Vertical transmission of caprine brucellosis was evident causing mild to moderate lesions in different organs. The samples of choice for isolation and identification of B. melitensis are stomach content as well as liver and spleen tissue.


Assuntos
Brucella melitensis , Brucelose/veterinária , Feto/microbiologia , Doenças das Cabras/microbiologia , Aborto Animal/microbiologia , Animais , Brucelose/microbiologia , Brucelose/patologia , Brucelose/transmissão , Feminino , Feto/patologia , Conteúdo Gastrointestinal/microbiologia , Doenças das Cabras/patologia , Doenças das Cabras/transmissão , Cabras , Transmissão Vertical de Doenças Infecciosas/veterinária , Gravidez , Complicações Infecciosas na Gravidez/microbiologia , Complicações Infecciosas na Gravidez/patologia , Complicações Infecciosas na Gravidez/veterinária , Natimorto/veterinária
17.
Infect Immun ; 89(2)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33139387

RESUMO

Listeria monocytogenes is a facultative Gram-positive intracellular bacterium that is capable of causing serious invasive infections in pregnant women, resulting in abortion, still-birth, and disseminated fetal infection. Previously, a clinical L. monocytogenes isolate, 07PF0776, was identified as having an enhanced ability to target cardiac tissue. This tissue tropism appeared to correlate with amino acid variations found within internalin B (InlB), a bacterial surface protein associated with host cell invasion. Given that the mammalian receptor bound by InlB, Met, is abundantly expressed by placental tissue, we assessed isolate 07PF0776 for its ability to be transmitted from mother to fetus. Pregnant Swiss Webster mice were infected on gestational day E13 via tail vein injection with the standard isolate 10403S, a noncardiotropic strain, or 07PF0776, the cardiac isolate. Pregnant mice infected with 07PF0776 exhibited significantly enhanced transmission of L. monocytogenes to placentas and fetuses compared to 10403S. Both bacterial burdens and the frequency of placental and fetal infection were increased in mice infected with the cardiac isolate. Strain 07PF0776 also exhibited an enhanced ability to invade Jar human trophoblast tissue culture cells in comparison to 10403S, and was found to have increased levels of InlB associated with the bacterial cell surface. Overexpression of surface InlB via genetic manipulation was sufficient to confer enhanced invasion of the placenta and fetus to both 10403S and 07PF0776. These data support a central role for surface InlB in promoting vertical transmission of L. monocytogenes.


Assuntos
Proteínas de Bactérias/fisiologia , Feto/fisiopatologia , Coração/fisiopatologia , Listeria monocytogenes/patogenicidade , Listeriose/transmissão , Proteínas de Membrana/fisiologia , Virulência/fisiologia , Adulto , Feminino , Feto/microbiologia , Coração/microbiologia , Humanos , Transmissão Vertical de Doenças Infecciosas , Masculino , Gravidez
18.
JCI Insight ; 5(21)2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33001863

RESUMO

Symbiotic microbial colonization through the establishment of the intestinal microbiome is critical to many intestinal functions, including nutrient metabolism, intestinal barrier integrity, and immune regulation. Recent studies suggest that education of intestinal immunity may be ongoing in utero. However, the drivers of this process are unknown. The microbiome and its byproducts are one potential source. Whether a fetal intestinal microbiome exists is controversial, and whether microbially derived metabolites are present in utero is unknown. Here, we aimed to determine whether bacterial DNA and microbially derived metabolites can be detected in second trimester human intestinal samples. Although we were unable to amplify bacterial DNA from fetal intestines, we report a fetal metabolomic intestinal profile with an abundance of bacterially derived and host-derived metabolites commonly produced in response to microbiota. Though we did not directly assess their source and function, we hypothesize that these microbial-associated metabolites either come from the maternal microbiome and are vertically transmitted to the fetus to prime the fetal immune system and prepare the gastrointestinal tract for postnatal microbial encounters or are produced locally by bacteria that were below our detection threshold.


Assuntos
Bactérias/metabolismo , Feto/metabolismo , Microbioma Gastrointestinal , Trato Gastrointestinal/metabolismo , Intestinos/fisiologia , Metaboloma , Adolescente , Bactérias/genética , Bactérias/isolamento & purificação , Criança , DNA Bacteriano/análise , DNA Bacteriano/genética , Feminino , Feto/microbiologia , Trato Gastrointestinal/microbiologia , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Intestinos/microbiologia , Masculino
19.
Sci Rep ; 10(1): 17712, 2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-33077862

RESUMO

Foetus sterility until parturition is under debate due to reports of microorganisms in the foetal environment and meconium. Sufficient controls to overcome sample contamination and provide direct evidence of microorganism viability in the pre-rectal gastrointestinal tract (GIT) have been lacking. We conducted molecular and culture-based analyses to investigate the presence of a microbiome in the foetal GIT of calves at 5, 6 and 7 months gestation, while controlling for contamination. The 5 components of the GIT (ruminal fluid, ruminal tissue, caecal fluid, caecal tissue and meconium) and amniotic fluid were found to contain a pioneer microbiome of distinct bacterial and archaeal communities. Bacterial and archaeal richness varied between GIT components. The dominant bacterial phyla in amniotic fluid differed to those in ruminal and caecal fluids and meconium. The lowest bacterial and archaeal abundances were associated with ruminal tissues. Viable bacteria unique to the ruminal fluids, which were not found in the controls from 5, 6 and 7 months gestation, were cultured, subcultured, sequenced and identified. We report that the foetal GIT is not sterile but is spatially colonised before birth by a pioneer microbiome.


Assuntos
Bovinos/embriologia , Feto/microbiologia , Microbiota , Animais , Archaea/classificação , Archaea/isolamento & purificação , Bactérias/classificação , Bactérias/isolamento & purificação , Trato Gastrointestinal/microbiologia
20.
mSphere ; 5(3)2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32376701

RESUMO

The prevailing paradigm in obstetrics has been the sterile womb hypothesis. However, some are asserting that the placenta, intra-amniotic environment, and fetus harbor microbial communities. The objective of this study was to determine whether the fetal and placental tissues of rhesus macaques harbor bacterial communities. Fetal, placental, and uterine wall samples were obtained from cesarean deliveries without labor (∼130/166 days gestation). The presence of bacteria in the fetal intestine and placenta was investigated through culture. The bacterial burden and profiles of the placenta, umbilical cord, and fetal brain, heart, liver, and colon were determined through quantitative real-time PCR and DNA sequencing. These data were compared with those of the uterine wall as well as to negative and positive technical controls. Bacterial cultures of fetal and placental tissues yielded only a single colony of Cutibacterium acnes This bacterium was detected at a low relative abundance (0.02%) in the 16S rRNA gene profile of the villous tree sample from which it was cultured, yet it was also identified in 12/29 background technical controls. The bacterial burden and profiles of fetal and placental tissues did not exceed or differ from those of background technical controls. By contrast, the bacterial burden and profiles of positive controls exceeded and differed from those of background controls. Among the macaque samples, distinct microbial signals were limited to the uterine wall. Therefore, using multiple modes of microbiologic inquiry, there was not consistent evidence of bacterial communities in the fetal and placental tissues of rhesus macaques.IMPORTANCE Microbial invasion of the amniotic cavity (i.e., intra-amniotic infection) has been causally linked to pregnancy complications, especially preterm birth. Therefore, if the placenta and the fetus are typically populated by low-biomass microbial communities, current understanding of the role of microbes in reproduction and pregnancy outcomes will need to be fundamentally reconsidered. Could these communities be of benefit by competitively excluding potential pathogens or priming the fetal immune system for the microbial bombardment it will experience upon delivery? If so, what properties (e.g., microbial load and community membership) of these microbial communities preclude versus promote intra-amniotic infection? Given the ramifications of the in utero colonization hypothesis, critical evaluation is required. In this study, using multiple modes of microbiologic inquiry (i.e., culture, quantitative real-time PCR [qPCR], and DNA sequencing) and controlling for potential background DNA contamination, we did not find consistent evidence for microbial communities in the placental and fetal tissues of rhesus macaques.


Assuntos
Feto/microbiologia , Microbiota , Placenta/microbiologia , Animais , Bactérias/classificação , Carga Bacteriana , Biópsia , Estudos Transversais , DNA Bacteriano/genética , Feminino , Feto/patologia , Macaca mulatta , Placenta/patologia , Gravidez , RNA Ribossômico 16S/genética , Útero/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...