Artigos de revistas sobre o tema "Mycobiote intestinal"

Siga este link para ver outros tipos de publicações sobre o tema: Mycobiote intestinal.

Crie uma referência precisa em APA, MLA, Chicago, Harvard, e outros estilos

Selecione um tipo de fonte:

Veja os 50 melhores artigos de revistas para estudos sobre o assunto "Mycobiote intestinal".

Ao lado de cada fonte na lista de referências, há um botão "Adicionar à bibliografia". Clique e geraremos automaticamente a citação bibliográfica do trabalho escolhido no estilo de citação de que você precisa: APA, MLA, Harvard, Chicago, Vancouver, etc.

Você também pode baixar o texto completo da publicação científica em formato .pdf e ler o resumo do trabalho online se estiver presente nos metadados.

Veja os artigos de revistas das mais diversas áreas científicas e compile uma bibliografia correta.

1

James, Steve A., Aimee Parker, Catherine Purse, Andrea Telatin, David Baker, Sandy Holmes, James Durham, Simon G. P. Funnell e Simon R. Carding. "The Cynomolgus Macaque Intestinal Mycobiome Is Dominated by the Kazachstania Genus and K. pintolopesii Species". Journal of Fungi 8, n.º 10 (8 de outubro de 2022): 1054. http://dx.doi.org/10.3390/jof8101054.

Texto completo da fonte
Resumo:
The cynomolgus macaque, Macaca fascicularis, is a non-human primate (NHP) widely used in biomedical research as its genetics, immunology and physiology are similar to those of humans. They may also be a useful model of the intestinal microbiome as their prokaryome resembles that of humans. However, beyond the prokaryome relatively little is known about other constituents of the macaque intestinal microbiome including the mycobiome. Here, we conducted a region-by-region taxonomic survey of the cynomolgus intestinal mycobiota, from duodenum to distal colon, of sixteen captive animals of differing age (from young to old). Using a high-throughput ITS1 amplicon sequencing-based approach, the cynomolgus gut mycobiome was dominated by fungi from the Ascomycota phylum. The budding yeast genus Kazachstania was most abundant, with the thermotolerant species K. pintolopesii highly prevalent, and the predominant species in both the small and large intestines. This is in marked contrast to humans, in which the intestinal mycobiota is characterised by other fungal genera including Candida and Saccharomyces, and Candida albicans. This study provides a comprehensive insight into the fungal communities present within the captive cynomolgus gut, and for the first time identifies K. pintolopesii as a candidate primate gut commensal.
Estilos ABNT, Harvard, Vancouver, APA, etc.
2

Chiappori, Federica, Francesca Anna Cupaioli, Arianna Consiglio, Noemi Di Nanni, Ettore Mosca, Vito Flavio Licciulli e Alessandra Mezzelani. "Analysis of Faecal Microbiota and Small ncRNAs in Autism: Detection of miRNAs and piRNAs with Possible Implications in Host–Gut Microbiota Cross-Talk". Nutrients 14, n.º 7 (23 de março de 2022): 1340. http://dx.doi.org/10.3390/nu14071340.

Texto completo da fonte
Resumo:
Intestinal microorganisms impact health by maintaining gut homeostasis and shaping the host immunity, while gut dysbiosis associates with many conditions, including autism, a complex neurodevelopmental disorder with multifactorial aetiology. In autism, gut dysbiosis correlates with symptom severity and is characterised by a reduced bacterial variability and a diminished beneficial commensal relationship. Microbiota can influence the expression of host microRNAs that, in turn, regulate the growth of intestinal bacteria by means of bidirectional host-gut microbiota cross-talk. We investigated possible interactions among intestinal microbes and between them and host transcriptional modulators in autism. To this purpose, we analysed, by “omics” technologies, faecal microbiome, mycobiome, and small non-coding-RNAs (particularly miRNAs and piRNAs) of children with autism and neurotypical development. Patients displayed gut dysbiosis related to a reduction of healthy gut micro- and mycobiota as well as up-regulated transcriptional modulators. The targets of dysregulated non-coding-RNAs are involved in intestinal permeability, inflammation, and autism. Furthermore, microbial families, underrepresented in patients, participate in the production of human essential metabolites negatively influencing the health condition. Here, we propose a novel approach to analyse faeces as a whole, and for the first time, we detected miRNAs and piRNAs in faecal samples of patients with autism.
Estilos ABNT, Harvard, Vancouver, APA, etc.
3

Li, Xin, Irina Leonardi, Alexa Semon, Itai Doron, Iris H. Gao, Gregory Garbe’s Putzel, Youngjun Kim et al. "Sensing Fungal Dysbiosis by Gut-Resident CX3CR1+ Mononuclear Phagocytes Aggravates Allergic Airway Disease". Journal of Immunology 202, n.º 1_Supplement (1 de maio de 2019): 191.3. http://dx.doi.org/10.4049/jimmunol.202.supp.191.3.

Texto completo da fonte
Resumo:
Abstract Sensing of the gut microbiota, including fungi, regulates mucosal immunity. Whether fungal sensing in the gut can influence immunity at other body sites is unknown. Here we show that fluconazole-induced gut fungal dysbiosis has persistent effects on allergic airway disease in a house dust mite challenge model. Mice with a defined community of bacteria, but lacking intestinal fungi were not susceptible to fluconazole-induced dysbiosis, while colonization with a fungal mixture recapitulated the detrimental effects. Gut-resident mononuclear phagocytes (MNPs) expressing the fractalkine receptor CX3CR1 were essential for the effect of gut fungal dysbiosis on peripheral immunity. However, how mycobiota influence immunity in gut distal sites is not well understood. We developed protocols for gut-targeted depletion of phagocytes to investigate the influence of fungi on gut-lung crosstalk. Depletion of CX3CR1+ MNPs or selective inhibition of Syk signaling downstream of fungal sensing in these cells ameliorated lung allergy. These results indicate that disruption of intestinal fungal communities can mediate gut-lung-directed immune crosstalk and aggravate disease severity through fungal sensing by gut-resident CX3CR1+ MNPs. Highlights Gut fungal dysbiosis persistently aggravates allergic airway disease (AAD) in mice. Gut colonization by commensal fungi is both required and sufficient to aggravate AAD in a mycobiome-free mouse model. Intestinal CX3CR1+ mononuclear phagocytes (MNPs) are essential for the systemic effects of gut fungal dysbiosis on AAD. Inhibition of Syk-mediated fungal sensing in intestinal CX3CR1+ MNPs ameliorates AAD.
Estilos ABNT, Harvard, Vancouver, APA, etc.
4

Moeller, Jesper B., Irina Leonardi, Anders Schlosser, Anne-Laure Flamar, Nicholas J. Bessman, Gregory Garbès Putzel, Theresa Thomsen et al. "Modulation of the fungal mycobiome is regulated by the chitin-binding receptor FIBCD1". Journal of Experimental Medicine 216, n.º 12 (10 de outubro de 2019): 2689–700. http://dx.doi.org/10.1084/jem.20182244.

Texto completo da fonte
Resumo:
Host–microbiota interactions are critical in regulating mammalian health and disease. In addition to bacteria, parasites, and viruses, beneficial communities of fungi (the mycobiome) are important modulators of immune- and tissue-homeostasis. Chitin is a major component of the fungal cell wall, and fibrinogen C containing domain 1 (FIBCD1) is a chitin-binding protein; however, the role of this molecule in influencing host–mycobiome interactions in vivo has never been examined. Here, we identify direct binding of FIBCD1 to intestinal-derived fungi and demonstrate that epithelial-specific expression of FIBCD1 results in significantly reduced fungal colonization and amelioration of fungal-driven intestinal inflammation. Collectively, these results identify FIBCD1 as a previously unrecognized microbial pattern recognition receptor through which intestinal epithelial cells can recognize and control fungal colonization, limit fungal dysbiosis, and dampen intestinal inflammation.
Estilos ABNT, Harvard, Vancouver, APA, etc.
5

Kanj, Amjad N., e Joseph H. Skalski. "Gut Mycobiome and Asthma". Journal of Fungi 10, n.º 3 (1 de março de 2024): 192. http://dx.doi.org/10.3390/jof10030192.

Texto completo da fonte
Resumo:
This review explores the ‘gut–lung axis’ in asthma with a focus on commensal fungal organisms. We explore how changes to the intestinal commensal fungal community composition alter lung immune function. We comprehensively review available studies that have profiled the composition of the gut mycobiome in adults and children with asthma, and discuss mechanisms of gut–lung interactions that have been described in animal models of asthma. Studies indicate that intestinal fungal dysbiosis, such as an increased abundance of certain fungi like Candida, can elevate the risk of asthma in children and exacerbate it in adults. This effect is mediated through various pathways: the host immune system’s sensing of dysbiosis via C-type lectin receptors (e.g., Dectin-2), the impact of pro-inflammatory fungal metabolites (e.g., 12,13-diHOME, prostaglandin E2), and the role of lung immune cells (e.g., group 2 innate lymphoid cells [ILC2], M2 macrophages). We also describe strategies for modulating the gut mycobiome as potential therapies for severe asthma. The review concludes by emphasizing the necessity for further research into the role of the gut mycobiome in asthma to deepen our understanding of these complex interactions.
Estilos ABNT, Harvard, Vancouver, APA, etc.
6

Zhang, Lin, Hui Zhan, Wenye Xu, Shuai Yan e Siew C. Ng. "The role of gut mycobiome in health and diseases". Therapeutic Advances in Gastroenterology 14 (janeiro de 2021): 175628482110471. http://dx.doi.org/10.1177/17562848211047130.

Texto completo da fonte
Resumo:
The gut microbiome comprised of microbes from multiple kingdoms, including bacteria, fungi, and viruses. Emerging evidence suggests that the intestinal fungi (the gut “mycobiome”) play an important role in host immunity and inflammation. Advances in next generation sequencing methods to study the fungi in fecal samples and mucosa tissues have expanded our understanding of gut fungi in intestinal homeostasis and systemic immunity in health and their contribution to different human diseases. In this review, the current status of gut mycobiome in health, early life, and different diseases including inflammatory bowel disease, colorectal cancer, and metabolic diseases were summarized.
Estilos ABNT, Harvard, Vancouver, APA, etc.
7

Bellotti, Ruben, Cornelia Speth, Timon E. Adolph, Cornelia Lass-Flörl, Maria Effenberger, Dietmar Öfner e Manuel Maglione. "Micro- and Mycobiota Dysbiosis in Pancreatic Ductal Adenocarcinoma Development". Cancers 13, n.º 14 (8 de julho de 2021): 3431. http://dx.doi.org/10.3390/cancers13143431.

Texto completo da fonte
Resumo:
Background: Dysbiosis of the intestinal flora has emerged as an oncogenic contributor in different malignancies. Recent findings suggest a crucial tumor-promoting role of micro- and mycobiome alterations also in the development of pancreatic ductal adenocarcinoma (PDAC). Methods: To summarize the current knowledge about this topic, a systematic literature search of articles published until October 2020 was performed in MEDLINE (PubMed). Results: An increasing number of publications describe associations between bacterial and fungal species and PDAC development. Despite the high inter-individual variability of the commensal flora, some studies identify specific microbial signatures in PDAC patients, including oral commensals like Porphyromonas gingivalis and Fusobacterium nucleatum or Gram-negative bacteria like Proteobacteria. The role of Helicobacter spp. remains unclear. Recent isolation of Malassezia globosa from PDAC tissue suggest also the mycobiota as a crucial player of tumorigenesis. Based on described molecular mechanisms and interactions between the pancreatic tissue and the immune system this review proposes a model of how the micro- and the mycobial dysbiosis could contribute to tumorigenesis in PDAC. Conclusions: The presence of micro- and mycobial dysbiosis in pancreatic tumor tissue opens a fascinating perspective on PDAC oncogenesis. Further studies will pave the way for novel tumor markers and treatment strategies.
Estilos ABNT, Harvard, Vancouver, APA, etc.
8

Tang, Jie, Iliyan D. Iliev, Jordan Brown, David M. Underhill e Vincent A. Funari. "Mycobiome: Approaches to analysis of intestinal fungi". Journal of Immunological Methods 421 (junho de 2015): 112–21. http://dx.doi.org/10.1016/j.jim.2015.04.004.

Texto completo da fonte
Estilos ABNT, Harvard, Vancouver, APA, etc.
9

Leonardi, Irina, Xin Li, Alexa Semon, Dalin Li, Itai Doron, Gregory Putzel, Agnieszka Bar et al. "CX3CR1+mononuclear phagocytes control immunity to intestinal fungi". Science 359, n.º 6372 (11 de janeiro de 2018): 232–36. http://dx.doi.org/10.1126/science.aao1503.

Texto completo da fonte
Resumo:
Intestinal fungi are an important component of the microbiota, and recent studies have unveiled their potential in modulating host immune homeostasis and inflammatory disease. Nonetheless, the mechanisms governing immunity to gut fungal communities (mycobiota) remain unknown. We identified CX3CR1+mononuclear phagocytes (MNPs) as being essential for the initiation of innate and adaptive immune responses to intestinal fungi. CX3CR1+MNPs express antifungal receptors and activate antifungal responses in a Syk-dependent manner. Genetic ablation of CX3CR1+MNPs in mice led to changes in gut fungal communities and to severe colitis that was rescued by antifungal treatment. In Crohn’s disease patients, a missense mutation in the gene encoding CX3CR1 was identified and found to be associated with impaired antifungal responses. These results unravel a role of CX3CR1+MNPs in mediating interactions between intestinal mycobiota and host immunity at steady state and during inflammatory disease.
Estilos ABNT, Harvard, Vancouver, APA, etc.
10

Xiang, Jun-Yan, Yan-Yu Chi, Jin-Xin Han, Ping Kong, Zehua Liang, Deli Wang, Hongyu Xiang e Qiuhong Xie. "Litchi chinensis seed prevents obesity and modulates the gut microbiota and mycobiota compositions in high-fat diet-induced obese zebrafish". Food & Function 13, n.º 5 (2022): 2832–45. http://dx.doi.org/10.1039/d1fo03991a.

Texto completo da fonte
Estilos ABNT, Harvard, Vancouver, APA, etc.
11

Reinoso, Samira, María Soledad Gutiérrez, Angélica Reyes-Jara, Magaly Toro, Katherine García, Guillermo Reyes, Wilfrido Argüello-Guevara, Milton Bohórquez-Cruz, Stanislaus Sonnenholzner e Paola Navarrete. "Feed Regime Slightly Modifies the Bacterial but Not the Fungal Communities in the Intestinal Mucosal Microbiota of Cobia Fish (Rachycentron canadum)". Microorganisms 11, n.º 9 (14 de setembro de 2023): 2315. http://dx.doi.org/10.3390/microorganisms11092315.

Texto completo da fonte
Resumo:
The bacterial community of the intestinal microbiota influences many host functions, and similar effects have been recently reported for the fungal community (mycobiota). Cobia is a tropical fish that has been studied for its potential in marine aquaculture. However, the study of its bacterial community has been underreported and the mycobiota has not been investigated. We analyzed the gut bacterial and fungal profile present in the intestinal mucosa of reared adult cobias fed two diets (frozen fish pieces (FFPs) and formulated feed (FF)) for 4 months by sequencing the 16S rRNA (V3-V4) and internal transcribed spacer-2 (ITS2) regions using Illumina NovaSeq 6000. No significant differences in the alpha diversity of the bacterial community were observed, which was dominated by the phyla Proteobacteria (~96%) and Firmicutes (~1%). Cobia fed FF showed higher abundance of 10 genera, mainly UCG-002 (Family Oscillospiraceae) and Faecalibacterium, compared to cobia fed FFPs, which showed higher abundance of 7 genera, mainly Methylobacterium-Methylorubrum and Cutibacterium. The inferred bacterial functions were related to metabolism, environmental information processing and cellular processes; and no differences were found between diets. In mycobiota, no differences were observed in the diversity and composition of cobia fed the two diets. The mycobiota was dominated by the phyla Ascomycota (~88%) and Basidiomycota (~11%). This is the first study to describe the gut bacterial and fungal communities in cobia reared under captive conditions and fed on different diets and to identify the genus Ascobulus as a new member of the core fish mycobiota.
Estilos ABNT, Harvard, Vancouver, APA, etc.
12

Van Thiel, I., T. Maasland, E. van Wassenaer, D. Hoekman, C. Spooren, T. Hakvoort, I. Admiraal et al. "P076 Abdominal pain severity for IBD in remission correlates with genetic clustering and enzymatic activity of faeces-derived Candida albicans strains". Journal of Crohn's and Colitis 17, Supplement_1 (30 de janeiro de 2023): i241—i242. http://dx.doi.org/10.1093/ecco-jcc/jjac190.0206.

Texto completo da fonte
Resumo:
Abstract Background Abdominal pain is a common occurrence for approximately 39% of patients with inflammatory bowel diseases in remission (quiescent IBD; qIBD). There is increasing evidence for a contributing role of the gastrointestinal fungal community (i.e., mycobiome) in relation to intestinal inflammation and irritable bowel syndrome (IBS). As such, abundance of Candida spp. and sub-species variation of Candida albicans were previously associated with IBD severity. In the current study, we aim to investigate mycobiome of patients with qIBD and qIBD with abdominal pain (qIBD–AP). Methods Patients with qIBD (defined as faecal calprotectin (FCP) ≤250 μg/g) with (n=91) or without (n=58) abdominal pain provided faecal samples. Abdominal pain was scored based on either the Irritable Bowel Syndrome Symptom Severity Scale (IBS–SSS) or Gastrointestinal Symptom Rating Score (GSRS) questionnaire. Faecal fungal communities were determined based on Internal Transcribed Spacer 1 (ITS1) sequencing. Cultivable yeasts from faecal samples were identified using Matrix Assisted Laser Deionization Time–of–Flight Mass Spectrometry. C. albicans strains (n=137 from 29 patients) were genotyped by ITS Sanger sequencing analysis and typing of seven microsatellite loci. Release of virulence-related enzymes (proteinase, phospholipase, lipase, esterase) by C. albicans was assessed through determination of precipitation zones on solid agar mediums containing enzyme-specific substrates. Results Descriptive mycobiota analysis revealed limited differences between compositions of qIBD and qIBD–AP patients, but a discriminative signature for abdominal pain was extracted using machine-based learning models. While ITS Sanger sequencing of faeces-derived C. albicans was insufficient to elucidate genetic variability, analysis of microsatellite loci revealed extensive variability and clustering into six clone clusters and a likely distinction between qIBD and qIBD-AP patients (Fig. 1). Phospholipase enzymatic activity of C. albicans strains correlated with GSRS–Abdominal Pain sub-score, as did proteinase activity with severity of abdominal pain according to the IBS–SSS (Fig. 2). Additionally, lipase activity inversely correlated with faecal calprotectin (Fig. 2). Conclusion The faecal gut mycobiome is associated with self–reported abdominal pain in patients with IBD in remission. This notion is based on both compositional and culture-dependent methods and specified clones of C. albicans may selectively contribute to abdominal pain for qIBD patients. This study opens further possibilities to investigate the role of faecal gut fungi in light of abdominal pain for qIBD-AP patients.
Estilos ABNT, Harvard, Vancouver, APA, etc.
13

Rolling, Thierry, Tobias M. Hohl e Bing Zhai. "Minority report: the intestinal mycobiota in systemic infections". Current Opinion in Microbiology 56 (agosto de 2020): 1–6. http://dx.doi.org/10.1016/j.mib.2020.05.004.

Texto completo da fonte
Estilos ABNT, Harvard, Vancouver, APA, etc.
14

Sciavilla, Piero, Francesco Strati, Monica Di Paola, Monica Modesto, Francesco Vitali, Duccio Cavalieri, Gian Maria Prati et al. "Gut microbiota profiles and characterization of cultivable fungal isolates in IBS patients". Applied Microbiology and Biotechnology 105, n.º 8 (abril de 2021): 3277–88. http://dx.doi.org/10.1007/s00253-021-11264-4.

Texto completo da fonte
Resumo:
Abstract Studies so far conducted on irritable bowel syndrome (IBS) have been focused mainly on the role of gut bacterial dysbiosis in modulating the intestinal permeability, inflammation, and motility, with consequences on the quality of life. Limited evidences showed a potential involvement of gut fungal communities. Here, the gut bacterial and fungal microbiota of a cohort of IBS patients have been characterized and compared with that of healthy subjects (HS). The IBS microbial community structure differed significantly compared to HS. In particular, we observed an enrichment of bacterial taxa involved in gut inflammation, such as Enterobacteriaceae, Streptococcus, Fusobacteria, Gemella, and Rothia, as well as depletion of health-promoting bacterial genera, such as Roseburia and Faecalibacterium. Gut microbial profiles in IBS patients differed also in accordance with constipation. Sequence analysis of the gut mycobiota showed enrichment of Saccharomycetes in IBS. Culturomics analysis of fungal isolates from feces showed enrichment of Candida spp. displaying from IBS a clonal expansion and a distinct genotypic profiles and different phenotypical features when compared to HS of Candida albicans isolates. Alongside the well-characterized gut bacterial dysbiosis in IBS, this study shed light on a yet poorly explored fungal component of the intestinal ecosystem, the gut mycobiota. Our results showed a differential fungal community in IBS compared to HS, suggesting potential for new insights on the involvement of the gut mycobiota in IBS. Key points • Comparison of gut microbiota and mycobiota between IBS and healthy subjects • Investigation of cultivable fungi in IBS and healthy subjects • Candida albicans isolates result more virulent in IBS subjects compared to healthy subjects
Estilos ABNT, Harvard, Vancouver, APA, etc.
15

Sadek, Ali, Bernard Taminiau, Georges Daube, Panagiotis Sapountzis, Frédérique Chaucheyras-Durand, Mathieu Castex, Françoise Coucheney e Djamel Drider. "Impact of Dietary Regime and Seasonality on Hindgut’s Mycobiota Diversity in Dairy Cows". Microorganisms 12, n.º 1 (31 de dezembro de 2023): 84. http://dx.doi.org/10.3390/microorganisms12010084.

Texto completo da fonte
Resumo:
We describe and discuss the intestinal mycobiota of dairy cows reared in France following variations in dietary regimes and two seasons. Two groups of 21 animals were followed over a summer and winter period, and another group of 28 animals was followed only during the same summer season. The summer diet was based on grazing supplemented with 3–5 kg/d of maize, grass silage and hay, while the winter diet consisted of 30% maize silage, 25% grass silage, 15% hay and 30% concentrate. A total of 69 DNA samples were extracted from the feces of these cows. Amplification and sequencing of the ITS2 region were used to assess mycobiota diversity. Analyses of alpha and beta diversity were performed and compared statistically. The mycobiota changed significantly from summer to winter conditions with a decrease in its diversity, richness and evenness parameters, while beta diversity analysis showed different mycobiota profiles. Of note, the Geotrichum operational taxonomic unit (OTU) was prevalent in the winter group, with a mean relative abundance (RA) of 65% of the total mycobiota. This Geotrichum OTU was also found in the summer group, but to a lesser extent (5%). In conclusion, a summer grazing diet allowed a higher fecal fungal diversity. These data show, for the first time, that a change in diet associated with seasonality plays a central role in shaping hindgut fungal diversity.
Estilos ABNT, Harvard, Vancouver, APA, etc.
16

Yadav, Meeta, Soham Ali, Rachel L. Shrode, Shailesh K. Shahi, Samantha N. Jensen, Jemmie Hoang, Samuel Cassidy et al. "Multiple sclerosis patients have an altered gut mycobiome and increased fungal to bacterial richness". PLOS ONE 17, n.º 4 (26 de abril de 2022): e0264556. http://dx.doi.org/10.1371/journal.pone.0264556.

Texto completo da fonte
Resumo:
Trillions of microbes such as bacteria, fungi, and viruses exist in the healthy human gut microbiome. Although gut bacterial dysbiosis has been extensively studied in multiple sclerosis (MS), the significance of the fungal microbiome (mycobiome) is an understudied and neglected part of the intestinal microbiome in MS. The aim of this study was to characterize the gut mycobiome of patients with relapsing-remitting multiple sclerosis (RRMS), compare it to healthy controls, and examine its association with changes in the bacterial microbiome. We characterized and compared the mycobiome of 20 RRMS patients and 33 healthy controls (HC) using Internal Transcribed Spacer 2 (ITS2) and compared mycobiome interactions with the bacterial microbiome using 16S rRNA sequencing. Our results demonstrate an altered mycobiome in RRMS patients compared with HC. RRMS patients showed an increased abundance of Basidiomycota and decreased Ascomycota at the phylum level with an increased abundance of Candida and Epicoccum genera along with a decreased abundance of Saccharomyces compared to HC. We also observed an increased ITS2/16S ratio, altered fungal and bacterial associations, and altered fungal functional profiles in MS patients compared to HC. This study demonstrates that RRMS patients had a distinct mycobiome with associated changes in the bacterial microbiome compared to HC. There is an increased fungal to bacterial ratio as well as more diverse fungal-bacterial interactions in RRMS patients compared to HC. Our study is the first step towards future studies in delineating the mechanisms through which the fungal microbiome can influence MS disease.
Estilos ABNT, Harvard, Vancouver, APA, etc.
17

Yadav, Meeta, Soham Ali, Rachel L. Shrode, Shailesh K. Shahi, Samantha N. Jensen, Jemmie Hoang, Samuel Cassidy et al. "Multiple sclerosis patients have an altered gut mycobiome and increased fungal to bacterial richness". PLOS ONE 17, n.º 4 (26 de abril de 2022): e0264556. http://dx.doi.org/10.1371/journal.pone.0264556.

Texto completo da fonte
Resumo:
Trillions of microbes such as bacteria, fungi, and viruses exist in the healthy human gut microbiome. Although gut bacterial dysbiosis has been extensively studied in multiple sclerosis (MS), the significance of the fungal microbiome (mycobiome) is an understudied and neglected part of the intestinal microbiome in MS. The aim of this study was to characterize the gut mycobiome of patients with relapsing-remitting multiple sclerosis (RRMS), compare it to healthy controls, and examine its association with changes in the bacterial microbiome. We characterized and compared the mycobiome of 20 RRMS patients and 33 healthy controls (HC) using Internal Transcribed Spacer 2 (ITS2) and compared mycobiome interactions with the bacterial microbiome using 16S rRNA sequencing. Our results demonstrate an altered mycobiome in RRMS patients compared with HC. RRMS patients showed an increased abundance of Basidiomycota and decreased Ascomycota at the phylum level with an increased abundance of Candida and Epicoccum genera along with a decreased abundance of Saccharomyces compared to HC. We also observed an increased ITS2/16S ratio, altered fungal and bacterial associations, and altered fungal functional profiles in MS patients compared to HC. This study demonstrates that RRMS patients had a distinct mycobiome with associated changes in the bacterial microbiome compared to HC. There is an increased fungal to bacterial ratio as well as more diverse fungal-bacterial interactions in RRMS patients compared to HC. Our study is the first step towards future studies in delineating the mechanisms through which the fungal microbiome can influence MS disease.
Estilos ABNT, Harvard, Vancouver, APA, etc.
18

Yin, Yue, Maermaer Tuohutaerbieke, Chengjie Feng, Xinjie Li, Yuqi Zhang, Qiang Xu, Jing Tu, Ence Yang, Qinghua Zou e Tao Shen. "Characterization of the Intestinal Fungal Microbiome in HIV and HCV Mono-Infected or Co-Infected Patients". Viruses 14, n.º 8 (18 de agosto de 2022): 1811. http://dx.doi.org/10.3390/v14081811.

Texto completo da fonte
Resumo:
Intestinal mycobiome dysbiosis plays an important role in the advancement of HIV- and HCV-infected patients. Co-infection with HCV is an important risk factor for exacerbating immune activation in HIV-infected patients, and gut fungal microbial dysbiosis plays an important role. However, no systematic study has been conducted on the intestinal fungal microbiome of HIV/HCV co-infected patients to date. Patients infected with HIV and HCV, either alone or in combination, and healthy volunteers were included. Stool samples were collected for fungal ITS sequencing and for further mycobiome statistical analysis. We found that the abundance of fungal species significantly decreased in the HIV/HCV co-infection group compared to in the healthy control group, while no significant differences were found in the mono-infection groups. Low-CD4 + T-cell patients in the HIV group and high-ALT-level patients in the HCV group were discovered to have a more chaotic fungal community. Furthermore, the opportunistic pathogenic fungal profiles and fungal inter-correlations in the co-infection group became less characteristic but more complicated than those in the mono-infection groups. Intestinal fungal dysregulation occurs in HIV- and HCV-infected patients, and this dysregulation is further complicated in HIV/HCV co-infected patients.
Estilos ABNT, Harvard, Vancouver, APA, etc.
19

Restrepo-Rivera, Lina Marcela, e Nora Cardona-Castro. "Micobioma: diversidad fúngica en el cuerpo humano". CES Medicina 35, n.º 2 (6 de julho de 2021): 113–25. http://dx.doi.org/10.21615/cesmedicina.5686.

Texto completo da fonte
Resumo:
Introducción: los hongos hacen parte de los microorganismos que se encuentran en el ser humano y que interactúan con bacterias, virus y archaeas. El equilibrio inter e intra-especies es importante para mantener la salud en los seres humanos. La mayoría de los estudios sobre la micobioma se han relacionado con estados de enfermedad causados por hongos, siendo de relevancia la exploración de las comunidades comensales en individuos sanos. Métodos: se realizó una búsqueda bibliográfica en PubMed, ScienceDirect, Scopus y Google Scholar, usando los términos mycobiome, intestinal fungi, skin mycobiome, vaginal mycobiome, fungal microbiome. Se incluyeron artículos desde 1996 hasta 2020, de revisión y de resultados de investigación en todos los idiomas. Resultados y discusión: existe variabilidad en las comunidades fúngicas en los sitios corporales según sus características intrínsecas y la relación con el medio ambiente. El estado de salud en los seres humanos puede estar influenciado por la densidad y diversidad fúngica, a diferencia de los individuos enfermos en quienes se evidencia una disminución en la diversidad y que se asocia con el oportunismo de agentes patógenos.
Estilos ABNT, Harvard, Vancouver, APA, etc.
20

Zhai, Bing, Mihaela Ola, Thierry Rolling, Nicholas L. Tosini, Sari Joshowitz, Eric R. Littmann, Luigi A. Amoretti et al. "High-resolution mycobiota analysis reveals dynamic intestinal translocation preceding invasive candidiasis". Nature Medicine 26, n.º 1 (janeiro de 2020): 59–64. http://dx.doi.org/10.1038/s41591-019-0709-7.

Texto completo da fonte
Estilos ABNT, Harvard, Vancouver, APA, etc.
21

Leonardi, Irina, e Iliyan Iliev. "3 MECHANSISMS OF INTESTINAL FUNGI RECOGNITION AND CONTROL". Inflammatory Bowel Diseases 26, Supplement_1 (janeiro de 2020): S41. http://dx.doi.org/10.1093/ibd/zaa010.107.

Texto completo da fonte
Resumo:
Abstract Background and Objectives Intestinal fungal communities are perturbed in several autoimmune diseases and have been shown to influence disease outcome. We have shown that intestinal resident CX3CR1+ mononuclear phagocytes (MNPs) can sense gut fungi and are crucial for the initiation of immune responses both locally and at distant sites. These results suggest that recognition of fungi by gut phagocytes might be involved in the pathogenesis of immune-related diseases such as IBD. Despite the identification of a few molecules involved in the recognition and immunity to intestinal fungi the cellular mechanisms governing the initiation and regulation of the mucosal immune responses to the mycobiota remain unknown. We sought to identify the functional role of distinct phagocytic subsets in the response to fungal communities in the gut during health and intestinal disease. Methods We used Candida albicans, the main opportunistic fungus found in IBD patients, as model fungal colonizer, with a focus on the mechanisms mediating the adaptive response. To elucidate the mechanisms and consequences of the recognition of fungi by phagocytic subsets in the lamina propria we used genetic models of depletion and deletion of specific subtypes of phagocytes. We further targeted fungal recognition and antigen presentation in phagocytes to investigate the mechanisms leading to the induction of adaptive anti-fungal responses. Results C. albicans colonization induced a consistent increase in Th17 cells in the intestinal mucosa that was decreased upon depletion of CX3CR1+ MNPs. Genetic depletion of CX3CR1+ MNPs in mice led to changes in gut fungal communities and to severe chemically induced intestinal inflammation that was rescued by antifungal treatment. Recognition of fungi through a C-type lectin/Syk pathways and antigen presentation via MHC-II were necessary for the induction of adaptive T cell in responses to C. albicans colonization. Conclusion Our work aims at defining the role of CX3CR1+ MNPs and cDCs in the initiation of anti-fungal immune responses in the intestine at the steady state. We have demonstrated the essential role of CX3CR1+ MNPs in the initiation of antifungal responses in the intestine at steady state and for the control of fungi at steady state and during inflammation. We have demonstrated the importance of CX3CR1+ MNPs in the control of the intestinal mycobiota.
Estilos ABNT, Harvard, Vancouver, APA, etc.
22

McAndrew, Morgan, Hannah Havran, Luca Di Martino, Mahmoud Ghannoum, Theresa Pizarro e Carlo De Salvo. "NOD2 PROMOTES HOST DEFENSE AND RECOVERY INDUCING MACROPHAGE-MEDIATED INNATE LYMPHOID CELLS TYPE 3 ACTIVATION FOLLOWING FUNGAL INFECTION IN EXPERIMENTAL COLITIS". Inflammatory Bowel Diseases 28, Supplement_1 (22 de janeiro de 2022): S59. http://dx.doi.org/10.1093/ibd/izac015.093.

Texto completo da fonte
Resumo:
Abstract Recent studies have documented the complexity of the intestinal fungal community (‘mycobiome’) in mice, and clinical and experimental observations have shown that the mycobiome influences both gut health and disease, e.g., inflammatory bowel disease (IBD). In fact, prior studies have shown that Crohn’s disease (CD) patients, compared to healthy controls, harbor higher levels of intestinal Candida tropicalis (Ct), which is the major fungal species detected in the colons of colitic mice after DSS challenge. More recently, Debaryomyces hansenii (also known as Candida famata) has been found in intestinal wounds of mice and inflamed mucosal tissues of CD patients. Moreover, proteins encoded by genes within IBD susceptibility loci, such as the pattern recognition receptor (PRR), NOD2, are known not only to recognize bacterial components, but also a fungal cell wall element, chitin. In fact, the role of PRRs in regulating immunity against intestinal fungi, and how fungi influence IBD remains poorly defined. We challenged DSS colitic WT and Nod2-/- mice with Ct 2 days before DSS administration and subsequently on day (d)0, 3 and 6. Our data confirms previous studies that Ct challenge does not exacerbate colitis in DSS-treated C57BL/6 wildtype (WT) mice, however, Ct-infected Nod2-/- mice possess a higher fungal burden and exhibit worse colitis symptoms, such as weight loss, decreased stool consistency, and presence of blood in stools, vs. Ct-infected WT mice, indicating an essential and protective role for NOD2 during colitis recovery after Ct challenge. Our results also show that Ct-infected Nod2-/- mice display a marked reduction in colonic Il22 and Il17, which are cytokines previously reported to be important in maintaining epithelial barrier integrity during DSS colitis, These data were confirmed in colons of Ct infected DSS challenged, ileitis-prone SAMP1/YitFc (SAMP) mice that were deficient in NOD2. Moreover, Our in vitro data show, a decrease in Il1b and Il23 in bone marrow-derived macrophages from SAMP Nod2-/- mice compared to WT after 2h of exposure to chitin. IL-17+ innate lymphoid cells (ILCs) are also known to control fungal burden during opportunistic fungal infections, and interestingly, our findings indicate that Ct infection of Nod2-/- vs. WT mice results in a decreased frequency of mesenteric lymph node–derived type 3 ILCs (ILC3s), suggesting that delay in fungal clearance and recovery in Nod2-/- mice may be due to the inability to mount protective type 3 immune responses. Taken together, the data so far collected suggests that NOD2 is essential to maintain gut mycobiome homeostasis and drives protective innate immune responses, via a macrophage-mediated ILC3 recruitment and IL-17 mechanism, by preventing the overgrowth of opportunistic fungi that may contribute to chronic intestinal inflammation, such as that observed in CD.
Estilos ABNT, Harvard, Vancouver, APA, etc.
23

De Salvo, C., H. Wargo, K. Vidmar, L. Di Martino, G. Mahabaleshwar, F. Cominelli, M. Ghannoum e T. Pizarro. "P063 Host-mycobiome interactions during the resolution of inflammation in experimental colitis". Journal of Crohn's and Colitis 17, Supplement_1 (30 de janeiro de 2023): i230—i232. http://dx.doi.org/10.1093/ecco-jcc/jjac190.0193.

Texto completo da fonte
Resumo:
Abstract Background Recent studies have documented the complexity of the intestinal fungal community (‘mycobiome’) in mice, and clinical and experimental observations have shown that the mycobiome influences both gut health and disease, e.g., IBD. In fact, prior studies have shown that CD patients, compared to healthy controls, harbor higher levels of intestinal Candida tropicalis (Ct), which is the major fungal species detected in the colons of colitic mice after dextran sodium sulfate (DSS) challenge. Moreover, proteins encoded by genes within IBD susceptibility loci, such as the pattern recognition receptor (PRR), NOD2, are known not only to recognize bacterial components, but also a fungal cell wall element, chitin. In fact, the role of PRRs in regulating immunity against intestinal fungi, and how fungi influence IBD remains poorly defined. Methods We challenged DSS colitic wildtype (WT) and Nod2-/- mice with Ct 2 days before DSS administration and subsequently on day 0, 3 and 6. Bone marrow-derived macrophages (BMDMs) from untreated mice were administered chitin in vitro. Results Our data confirms previous studies that Ct challenge does not exacerbate colitis in DSS-treated C57BL/6 WT mice, however, Ct-infected Nod2-/- mice possess a higher fungal burden and exhibit worse colitis symptoms, such as weight loss, decreased stool consistency, and presence of blood in stools, vs. Ct-infected WT mice, indicating an essential and protective role for NOD2 during colitis recovery after Ct challenge. Our results also show that Ct-infected Nod2-/- mice display a marked reduction in colonic Il22 and Il17, which are cytokines previously reported to be important in maintaining epithelial barrier integrity during DSS colitis. These data were confirmed in colons of Ct infected DSS challenged, ileitis prone SAMP1/YitFc (SAMP) mice NOD2 deficient. Moreover, our in vitro data show, a decrease in Il1ß and Il23 in BMDM from SAMP Nod2-/- mice compared to WT after 2h of exposure to chitin. IL-17+ innate lymphoid cells (ILCs) are also known to control fungal burden during opportunistic fungal infections, and interestingly, our findings indicate that Ct infection of Nod2-/- vs. WT mice results in a decreased frequency of mesenteric lymph node–derived type 3 ILCs (ILC3s), suggesting that delay in fungal clearance and recovery in Nod2-/-mice may be due to the inability to mount protective type 3 immune responses. Conclusion Taken together, the data collected suggests that NOD2 is essential to maintain gut mycobiome homeostasis and drives protective innate immune responses, via a macrophage-mediated ILC3 recruitment and IL-17 mechanism, by preventing the overgrowth of opportunistic fungi that may contribute to chronic intestinal inflammation, such as that observed in CD.
Estilos ABNT, Harvard, Vancouver, APA, etc.
24

Meineri, Giorgia, Elisa Martello, David Atuahene, Silvia Miretti, Bruno Stefanon, Misa Sandri, Ilaria Biasato, Maria Rita Corvaglia, Ilario Ferrocino e Luca Simone Cocolin. "Effects of Saccharomyces boulardii Supplementation on Nutritional Status, Fecal Parameters, Microbiota, and Mycobiota in Breeding Adult Dogs". Veterinary Sciences 9, n.º 8 (28 de julho de 2022): 389. http://dx.doi.org/10.3390/vetsci9080389.

Texto completo da fonte
Resumo:
The aim of this study was to evaluate the effect of the administration of Saccharomyces boulardii on the nutritional, immunological, inflammatory, and stress status and on the composition of the gut microbiota and mycobiota in healthy adult dogs. A total of 25 American Staffordshire Terrier dogs were selected and randomly assigned to two groups: control (CTR, n = 12) and treated (TRT, n = 13) groups. No significant differences were found between the two groups regarding body weight, body condition score, and fecal score. No significant differences in microbiota/mycobiota, short chain fatty acids, indole/skatole, histamine, zonulin, or lactoferrin were detected. Indeed, supplementation with S. boulardii significantly decreased fecal calprotectin Immunoglobulin A, indicating an improvement in the gut well-being. Interestingly, fecal cortisol significantly decreased in dogs belonging to the TRT group compared to the CTR, suggesting both an improvement of the intestinal status and a reduction of stress, a common condition affecting animals managed in a breeding environment.
Estilos ABNT, Harvard, Vancouver, APA, etc.
25

Valderrama, Benjamín, José J. Ruiz, María Soledad Gutiérrez, Katherine Alveal, Mario Caruffo, Marcia Oliva, Héctor Flores et al. "Cultivable Yeast Microbiota from the Marine Fish Species Genypterus chilensis and Seriolella violacea". Journal of Fungi 7, n.º 7 (28 de junho de 2021): 515. http://dx.doi.org/10.3390/jof7070515.

Texto completo da fonte
Resumo:
Because of its outstanding biological and industrial importance, many efforts have been made to characterize the mycobiota of new environments and their biochemical and biotechnological potentials. Gut mycobiota can be a source of novel yeasts with the potential to be used as probiotics or have industrial applications. In this work, we characterized two as-yet unexplored yeast communities from the intestinal content of the cultured marine Chilean fishes Genypterus chilensis (G. chilensis) and Seriolella violacea (S. violacea). Yeasts were isolated through culture, identified by sequencing their ITS region, and characterized their enzymatic profile with API®ZYM. Rhodotorula mucilaginosa was identified in both fish species. For the first time, Candida palmioleophila, Candida pseudorugosa, Cystobasidium slooffiae, and a member of the Yamadazyma genus were also identified and described as part of the normal fish gut–microbiota. Furthermore, the diverse enzymatic profile exhibited by some of these isolates suggests that it may be possible to develop novel applications for them, such as new probiotics and other biotechnological applications.
Estilos ABNT, Harvard, Vancouver, APA, etc.
26

García‐Gamboa, Ricardo, Manuel R. Kirchmayr, Misael Sebastian Gradilla‐Hernández, Vicente Pérez‐Brocal, Andrés Moya e Marisela González‐Avila. "The intestinal mycobiota and its relationship with overweight, obesity and nutritional aspects". Journal of Human Nutrition and Dietetics 34, n.º 4 (15 de fevereiro de 2021): 645–55. http://dx.doi.org/10.1111/jhn.12864.

Texto completo da fonte
Estilos ABNT, Harvard, Vancouver, APA, etc.
27

Gutierrez, Mackenzie W., e Marie-Claire Arrieta. "The intestinal mycobiome as a determinant of host immune and metabolic health". Current Opinion in Microbiology 62 (agosto de 2021): 8–13. http://dx.doi.org/10.1016/j.mib.2021.04.004.

Texto completo da fonte
Estilos ABNT, Harvard, Vancouver, APA, etc.
28

Guan, Jun, Shao-ze Liu, Zhao-fen Lin, Wen-fang Li, Xue-feng Liu e De-chang Chen. "Impact of imipenem treatment on colonic mycobiota in rats with double-hit sepsis". Chinese Medical Journal 126, n.º 10 (20 de maio de 2013): 1850–54. http://dx.doi.org/10.3760/cma.j.issn.0366-6999.20130201.

Texto completo da fonte
Resumo:
Background Broad-spectrum antibiotic administration promotes intestinal colonization of exogenous fungal pathogens in healthy animals and has been recognized as one of the risk factors of invasive fungal infection in clinical settings. It is unclear whether broad-spectrum antibiotic treatment would change the intestinal mycobiota without exogenous fungal challenge in the context of sepsis. Methods We established a rat model of double-hit sepsis using burn injury and endotoxin challenge. Rats with burn injury or double-hit sepsis received imipenem treatment for 3 days or 9 days, and their colon contents were sampled for selective fungal culture and isolation counts. Results Imipenem treatment promoted the overgrowth of the commensal fungus Geotrichum capitatum in rats with burn injury. Imipenem treatment also promoted colon colonization by exogenous fungi in rats with burn injury and double-hit sepsis, including Trichosporon cutaneum, Candida albicans, Candida krusei, and Candida glabrata. A longer duration of imipenem treatment had a stronger impact on colon colonization by exogenous fungi. Conclusion Imipenem treatment facilitates the overgrowth of commensal fungi and colonization by exogenous, potentially pathogenic fungi in the colons of rats with burn injury or double-hit sepsis.
Estilos ABNT, Harvard, Vancouver, APA, etc.
29

Reisdorf, Shannon, Hemamalini Bommiasamy, Michelle West, David Serody, Danny Bruce, Benjamin G. Vincent, James Coghill e Jonathan S. Serody. "Effect of Anti-Fungal Prophylaxis on the Fungal Mycobiome after Allogeneic Bone Marrow Transplantation". Blood 126, n.º 23 (3 de dezembro de 2015): 4290. http://dx.doi.org/10.1182/blood.v126.23.4290.4290.

Texto completo da fonte
Resumo:
Abstract Background: Intestinal bacteria shape the immune system and have been shown to contribute to GI tract inflammation. Modification of the lower GI tract bacterial microbiota has been correlated with decreased acute graft-versus-host disease (aGvHD) [1]. Additionally, decreased intestinal bacterial diversity has been associated with increased nonrelapse mortality following allogeneic stem cell transplantation (alloSCT), largely due to an association with severe aGvHD[2]. However, the role of the fungal mycobiome during aGvHD has been largely ignored. Antifungal prophylaxis with fluconazole has been found to decrease long-term transplant mortality following alloSCT. We wanted to determine (1) effect of anti-fungal prophylaxis on the fungal mycobiome and (2) whether enteric fungi contributes to aGvHD by promoting a pro-inflammatory environment in the GI tract, similar to certain bacteria. Methods: B6D2 recipient mice were lethally irradiated, and the following day, recipient mice received 3 x106 T cell depleted bone marrow cells and 4 x 106 naïve T cells from B6 donor mice. Recipient mice were followed for survival and scored for clinical GvHD using a standard scoring system. Mice were treated daily with the antifungal agent fluconazole at 5mg/kg/day. Stool samples were collected on days 0, 7, and 14 post-transplant, and DNA was isolated from stool. The fungal ribosomal internal transcribed spacer region (specifically ITS-1) was amplified by PCR and sequencing libraries generated using Illumina chemistry and evaluated on a MiSeq sequencer. Additionally, real-time PCR was performed on DNA from stool samples to quantitate fungi in the colon. Colons were harvested, immune cells isolated and evaluated for flow cytometric analysis after bone marrow transplantation. Intestinal permeability was assessed by oral administration of FITC-labeled dextran 4000, followed by peripheral blood collection to assess plasma FITC-dextran levels. Results: In B6D2 mice, the most common fungal species found in the colon before and after bone marrow transplantation was Candida tropicalis. We found a 50-75% decrease in the quantity of fungi present in the colon in fluconazole treated mice. However there was no alteration in the percentages of fungi found in the GI tract indicating that anti-fungal therapy alter the quantity and not the quality of the fungal microbiome. Specifically, we did not find that the administration of fluzonazole was associated with an increase in fluconazole-resistant Candida species such as C kruseii or with fungi intrinsically resistant to fluconazole such as Aspergillus organisms. While there was no difference in clinical scoring of GVHD or weight loss in fluconazole treated mice, there was a modest improvement in overall survival in the treated group. We found no differences in IFN-γ or IL-17 production by colonic T cells from mice treated with fluconazole as compared to control mice evaluated 14 days after transplant. Additionally, the percentage of Foxp3+ T cells was also similar between treated and control mice. We also found no change in aGvHD mediated intestinal permeability suggesting that fluconazole treatment has no effect on epithelial barrier function. Conclusion: These studies demonstrate that fluconazole decreases total colonic fungi without altering the relative frequency of fungal organisms and is not associated in mice with an increase in the presence of fluconazole-resistant fungi. Fluconazole treatment was associated with a modest improvement in overall survival. However, no differences were found in the percentage/number of donor TH1, TH17 or Treg cells in the colon. These data suggest that, in the B6->B6D2 model, decreasing fungal burden in the colon is associated with a modest improvement in overall survival without substantially altering the generation of pro-inflammatory T cells or Tregs. 1. Jenq RR et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J Exp Med. 2012 May 7;209(5):903-11. doi: 10.1084/jem.20112408. Epub 2012 Apr 30. 2. Taur Y, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014 Aug 14;124(7):1174-82. doi: 10.1182/blood -2014-02-554725. Epub 2014 Jun 17. Disclosures No relevant conflicts of interest to declare.
Estilos ABNT, Harvard, Vancouver, APA, etc.
30

Syromyatnikov, M. Yu, S. V. Shabunin, E. Yu Nesterova, M. I. Gladkikh, Yu D. Smirnova, I. Yu Burakova, P. D. Morozova, M. V. Gryaznova e E. V. Mikhaylov. "STUDY OF THE DIVERSITY OF FUNGAL MICROORGANISMS IN THE GUT OF SWINE WITH DIFFERENT FEED CONVERSION RATE". Transactions of the educational establishment “Vitebsk the Order of “the Badge of Honor” State Academy of Veterinary Medicine 59, n.º 4 (2023): 85–89. http://dx.doi.org/10.52368/2078-0109-2023-59-4-85-89.

Texto completo da fonte
Resumo:
The paper presents the results of study on high-throughput sequencing of the fungal microbiome in the intesti-nal tract of swine, as well as an assessment of the influence of the feed conversion rate on the biodiversity of the fun-gal community of the intestinal tract of animals. Bioinformatic analysis of nucleotide sequences revealed 3 bacterial phyla in the gut of swine, including Ascomycota, Basidiomycota and Microsporidia. The dominant phyla were Ascomycota and Basidiomycota. The most numerous fungi at the family level were representatives of Schizosaccharomycetaceae. The most abundant species in the intestinal microbiome of swine was Schizosaccharomyces pombe (the relative abundance for the two study groups was 9.24 ± 1.49). In the second place in prevalence was the species Colletotrichum higginsianum (relative abundance for group A: 5.44±0.98, for group B 5.97±0.76) and Thermothielavioides terrestris relative abundance for group A: 3.96±0, 67, for group B 4.37±0.46). A comparative bioinformatic analysis of the intestinal mycobiome of two groups of swine with different feed conversion rates did not reveal significant differences at the level of phylum, family and species. The results demonstrate the need for further detailed study of this issue.
Estilos ABNT, Harvard, Vancouver, APA, etc.
31

Mehal, Wajahat Z., e Robert F. Schwabe. "A disease-promoting role of the intestinal mycobiome in non-alcoholic fatty liver disease". Journal of Hepatology 76, n.º 4 (abril de 2022): 765–67. http://dx.doi.org/10.1016/j.jhep.2021.12.035.

Texto completo da fonte
Estilos ABNT, Harvard, Vancouver, APA, etc.
32

Kaźmierczak-Siedlecka, Karolina, Aleš Dvořák, Marcin Folwarski, Agnieszka Daca, Katarzyna Przewłócka e Wojciech Makarewicz. "Fungal Gut Microbiota Dysbiosis and Its Role in Colorectal, Oral, and Pancreatic Carcinogenesis". Cancers 12, n.º 5 (22 de maio de 2020): 1326. http://dx.doi.org/10.3390/cancers12051326.

Texto completo da fonte
Resumo:
The association between bacterial as well as viral gut microbiota imbalance and carcinogenesis has been intensively analysed in many studies; nevertheless, the role of fungal gut microbiota (mycobiota) in colorectal, oral, and pancreatic cancer development is relatively new and undiscovered field due to low abundance of intestinal fungi as well as lack of well-characterized reference genomes. Several specific fungi amounts are increased in colorectal cancer patients; moreover, it was observed that the disease stage is strongly related to the fungal microbiota profile; thus, it may be used as a potential diagnostic biomarker for adenomas. Candida albicans, which is the major microbe contributing to oral cancer development, may promote carcinogenesis via several mechanisms, mainly triggering inflammation. Early detection of pancreatic cancer provides the opportunity to improve survival rate, therefore, there is a need to conduct further studies regarding the role of fungal microbiota as a potential prognostic tool to diagnose this cancer at early stage. Additionally, growing attention towards the characterization of mycobiota may contribute to improve the efficiency of therapeutic methods used to alter the composition and activity of gut microbiota. The administration of Saccharomyces boulardii in oncology, mainly in immunocompromised and/or critically ill patients, is still controversial.
Estilos ABNT, Harvard, Vancouver, APA, etc.
33

Cieplińska, Katarzyna, Magdalena Gajęcka, Michał Dąbrowski, Anna Rykaczewska, Sylwia Lisieska-Żołnierczyk, Maria Bulińska, Łukasz Zielonka e Maciej T. Gajęcki. "Time-Dependent Changes in the Intestinal Microbiome of Gilts Exposed to Low Zearalenone Doses". Toxins 11, n.º 5 (24 de maio de 2019): 296. http://dx.doi.org/10.3390/toxins11050296.

Texto completo da fonte
Resumo:
Zearalenone is a frequent contaminant of cereals and their by-products in regions with a temperate climate. This toxic molecule is produced naturally by Fusarium fungi in crops. The aim of this study was to determine the influence of low zearalenone doses (LOAEL, NOAEL and MABEL) on the intestinal microbiome of gilts on different days of exposure (days 7, 21 and 42). Intestinal contents were sampled from the duodenal cap, the third part of the duodenum, jejunum, caecum and the descending colon. The experiment was performed on 60 clinically healthy gilts with average BW of 14.5 ± 2 kg, divided into three experimental groups and a control group. Group ZEN5 animals were orally administered ZEN at 5 μg /kg BW, group ZEN10—10 μg ZEN/kg BW and group ZEN15—15 µg ZEN/kg BW. Five gilts from every group were euthanized on analytical dates 1, 2 and 3. Differences in the log values of microbial counts, mainly Escherichia coli and Enterococcus faecalis, were observed between the proximal and distal segments of the intestinal tract on different analytical dates as well as in the entire intestinal tract. Zearalenone affected the colony counts of intestinal microbiota rather than microbiome diversity, and its effect was greatest in groups ZEN10 and ZEN15. Microbial colony counts were similar in groups ZEN5 and C. In the analysed mycobiome, ZEN exerted a stimulatory effect on the log values of yeast and mould counts in all intestinal segments, in particular in the colon, and the greatest increase was noted on the first analytical date.
Estilos ABNT, Harvard, Vancouver, APA, etc.
34

Foster, M. Lauren, Scot E. Dowd, Christine Stephenson, Jörg M. Steiner e Jan S. Suchodolski. "Characterization of the Fungal Microbiome (Mycobiome) in Fecal Samples from Dogs". Veterinary Medicine International 2013 (2013): 1–8. http://dx.doi.org/10.1155/2013/658373.

Texto completo da fonte
Resumo:
The prevalence and phylogenetic description of fungal organisms and their role as part of the intestinal ecosystem have not yet been studied extensively in dogs. This study evaluated the fungal microbiome of 19 dogs (12 healthy dogs and 7 dogs with acute diarrhea) using fungal tag-encoded FLX-Titanium amplicon pyrosequencing. Five distinct fungal phyla were identified, withAscomycota(medians: 97.9% of obtained sequences in healthy dogs and 98.2% in diseased dogs) andBasidiomycota(median 1.0% in healthy dogs and median 0.5% in diseased dogs) being the most abundant fungal phyla. A total of 219 fungal genera were identified across all 19 dogs with a median (range) of 28 (4–69) genera per sample.Candidawas the most abundant genus found in both the diseased dogs (median: 1.9%, range: 0.2%–38.5% of sequences) and healthy dogs (median: 5.2%, range: 0.0%–63.1% of sequences).Candida natalensiswas the most frequently identified species. No significant differences were observed in the relative proportions of fungal communities between healthy and diseased dogs. In conclusion, fecal samples of healthy dogs and dogs with acute diarrhea harbor various fungal genera, and their role in gastrointestinal health and disease warrants further studies.
Estilos ABNT, Harvard, Vancouver, APA, etc.
35

Meijer, Sofie, Elena Pasquinelli, Sonia Renzi, Shahram Lavasani, Mehrnaz Nouri, Lena Erlandsson, Duccio Cavalieri e Stefan R. Hansson. "Gut Micro- and Mycobiota in Preeclampsia: Bacterial Composition Differences Suggest Role in Pathophysiology". Biomolecules 13, n.º 2 (10 de fevereiro de 2023): 346. http://dx.doi.org/10.3390/biom13020346.

Texto completo da fonte
Resumo:
Preeclampsia is a severe pregnancy-related inflammatory disease without an effective treatment. The pathophysiology remains partly unknown. However, an increased inflammatory response and oxidative stress are part of the maternal systemic reaction. Recent data have suggested that dysbiosis of the gut microbiome plays a role in preeclampsia as well as other inflammatory diseases. However, dysbiosis in preeclampsia has not been studied in a Scandinavian population. Furthermore, although the fungal flora may also have anti-inflammatory properties, it has never been studied in preeclampsia. We included 25 preeclamptic and 29 healthy third-trimester women for the ITS and 16S sequencing of fungal and bacterial microbiota, respectively. Calprotectin was measured to assess systemic and intestinal inflammatory responses. The fungal diversity differed with BMI and gestational length, suggesting a link between fungi and the immune changes seen in pregnancy. An LEfSe analysis showed 18 significantly differentially abundant bacterial taxa in PE, including enriched Bacteroidetes and depleted Verrucomicrobia and Syntergistota at the phylum level and depleted Akkermansia at the genus level, suggesting a role in the pathophysiology of PE.
Estilos ABNT, Harvard, Vancouver, APA, etc.
36

Thiele Orberg, Erik, Elisabeth Meedt, Andreas Hiergeist, Jinling Xue, Sakhila Ghimire, Melanie Tiefgraber, Tina Eismann et al. "Longitudinal Analysis of Gut Bacteriome, Mycobiome, Virome and Metabolome in Allogeneic Stem Cell Transplantation Reveals Susceptibility for Acute Graft-Versus-Host Disease". Blood 138, Supplement 1 (5 de novembro de 2021): 332. http://dx.doi.org/10.1182/blood-2021-153026.

Texto completo da fonte
Resumo:
Abstract The intestinal microbiome directly effects outcomes in patients undergoing allogeneic stem cell transplantation (ASCT) (Peled et al., N Engl J Med, 2020). Recent developments in microbiome research allow us to look beyond bacterial communities at the composition of the intestinal mycobiome and virome. However, only few studies have addressed these fungal (Zhang et al., Nat Commun, 2021; Legoff et al., Nat Med, 2017) and viral communities (Van Der Velden et al., Biol Blood Marrow Transplant, 2013) in the ASCT setting. Loss of bacterial diversity and domination of single taxa was linked to increased incidence of graft-versus-host-disease (aGvHD) and mortality. However, the functional consequences of intestinal dysbiosis remain poorly understood. Recently, microbial-derived metabolites such as short-chain fatty acids (SCFAs), indoles and secondary bile acids (BA) have been proposed to explain how microbiota exert effects on epithelial and immune responses and shown to protect from GVHD in animal models. To assemble a complete picture of the phylogenetic and metabolic changes which occur as patients undergo ASCT we designed a multiomics approach and identified patterns in bacteriome, mycobiome, virome and metabolome dynamics in relation to clinical factors and GvHD. 353 stool samples from 80 patients undergoing ASCT were obtained at two different transplantation centers (Munich and Regensburg) at defined timepoints before and after transplantation. Changes in metabolite composition were evaluated via mass spectrometry using a targeted metabolomic profiling approach, accompanied by bacterial 16S rDNA-, fungal 18S-ITS- and deep virome sequencing. Results were linked among each other and with clinical metadata such as GvHD, response to steroids, antimicrobial therapy and mortality. Longitudinal profiling of ASCT patients revealed a significant loss of bacterial, fungal and viral diversity following ASCT. At both centers, the effect was most pronounced in the early post-transplant period. Patients that developed GI-GVHD had the lowest diversity scores (Panel 1). GVHD patient samples were dominated by Enteroccocus and Candida, and loss of Caudovirales species, indicative of deep dysbiosis effecting all intestinal communities. ASCT had a strong impact on microbiome function and their ability to produce metabolites. We assayed each sample for production of metabolites with reported protective effects: SCFAs (acetate, butyrate and propionate), indoles (indole-3-carboxylaldehyde, ICA) and BAs (deoxycholic and lithocholic acid). When admitted to the transplantation unit (timepoint "conditioning") most patients had rich metabolite expression profiles. In the early post-transplant period, metabolite levels became progressively more depleted. GVHD patients displayed a nearly complete loss of metabolites (Panel 2a). Importantly, levels of the metabolite ICA at early timepoints could predict the occurrence of GVHD, leading us to consider prophylactic administration in future studies. We performed beta diversity analysis to identify clinical factors that had the highest impact on microbial communities. As expected, we noted that administration of broad-spectrum antibiotics had a profound impact of bacteria, fungi and viruses; but also had a significant negative effect on metabolites levels. Lastly, we report on a case of successful treatment of severe GI-GVHD with fecal microbiota transplant. Response to treatment was characterized by the recipient adopting the FMT donor's rich metabolite expression profile (Panel 2b) . To our knowledge, this is the first multicenter study that combines bacteriome, mycobiome, virome and metabolome analyses a longitudinal fashion in matched patient cohorts. Our results are consistent with previous findings that there are major alterations in gut microbiome and metabolite composition after ASCT. We expand upon those findings by characterizing the time course of phylogenetic alterations and their functional consequences. Those alterations play an important role in GvHD pathogenesis and might serve as potential biomarkers to identify patients at high risk for developing lethal GvHD early on. Our results highlight two strategies for targeted microbiome-based interventions: (1) preventing loss of microbial communities by restrictive use of antibiotics and (2) prophylactic administration of metabolite cocktails. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
Estilos ABNT, Harvard, Vancouver, APA, etc.
37

De Jesus, Magdia, Emily Rochac-Argueta e Heather Gallagher. "Intestinal Regulation of C. albicans by PP Langerin+ DCs". Journal of Immunology 198, n.º 1_Supplement (1 de maio de 2017): 62.6. http://dx.doi.org/10.4049/jimmunol.198.supp.62.6.

Texto completo da fonte
Resumo:
Abstract As the largest mucosal compartment, the human gastrointestinal tract (GI) is highly colonized with a complex microbial ecosystem that includes fungal organisms. Although fewer in numbers and less extensively studied in the context of the intestinal mucosa, it is unclear whether commensal members such as Candida species play a role in intestinal mucosal immunity. Recent studies have suggested that Candida sp. in the intestinal mucosa are associated with a number of human diseases such as gastric ulcers, Crohn’s disease and Hirschsprung-associated enterocolitis. We have recently identified, a specific non-macrophage dendritic cell (DC) population within intestinal Peyer’s patches (PP) that expresses the C-type lectin receptor (CLR)-Langerin and are able to specifically uptake Candida albicans, Candida tropicalis and yeast purified β-1,3-glucan particles (GPs). We hypothesize that Langerin+ DCs are central in the sampling, presentation and regulation of C. albicans within PPs. To address this hypothesis, we are currently defining the role of Langerin+ DCs in the processing of Candida sp. after uptake using the Langerin-DTR- EGFP mouse model as well as the Langerin-DTR- EGFP-DSS model. We are also characterizing the local gene expression profiles of PP B and T- lymphocytes as well as Langerin+ DCs in response to C. albicans. Additionally, we are investigating whether Langerin+ DCs are a migratory cell subset that not only transports C. albicans near germinal centers but can also take them to tolerogenic sites such as the MLN. These studies will advance our knowledge of how commensal members that are also part of the “mycobiome” are sampled, what mucosal immune responses are elicited and how these fungal organisms are tolerated.
Estilos ABNT, Harvard, Vancouver, APA, etc.
38

Maas, Evy, John Penders e Koen Venema. "Fungal-Bacterial Interactions in the Human Gut of Healthy Individuals". Journal of Fungi 9, n.º 2 (19 de janeiro de 2023): 139. http://dx.doi.org/10.3390/jof9020139.

Texto completo da fonte
Resumo:
Most studies of the microbiota in the human gut focus on the bacterial part, but increasing information shows that intestinal fungi are also important for maintaining health. This can be either by directly influencing the host or by indirectly influencing the gut bacteria that link to host health. Studies of fungal communities in large cohorts are scarce; therefore, this study aims at obtaining more insight into the mycobiome of healthy individuals and how this mycobiome interacts with the bacterial component of the microbiome. For this purpose, ITS2 and 16S rRNA gene amplicon sequencing was performed on fecal samples from 163 individuals which were available from two separate studies to analyze the fungal and bacterial microbiome, respectively, as well as the cross-kingdom interactions. The results showed a much lower fungal, as compared to bacterial, diversity. Ascomycota and Basidiomycota were the dominant fungal phyla across all the samples, but levels varied enormously between individuals. The ten most abundant fungal genera were Saccharomyces, Candida, Dipodascus, Aureobasidium, Penicillium, Hanseniaspora, Agaricus, Debaryomyces, Aspergillus, and Pichia, and here also extensive inter-individual variation was observed. Correlations were made between bacteria and fungi, and only positive correlations were observed. One of the correlations was between Malassezia restricta and the genus Bacteroides, which have both been previously described as alleviated in IBD. Most of the other correlations found were with fungi that are not known as gut colonizers but originate from food and the environment. To further investigate the importance of the observed correlations found, more research is needed to discriminate between gut colonizers and transient species.
Estilos ABNT, Harvard, Vancouver, APA, etc.
39

Geng, Zixiang, Xiaoli Nie, Lele Ling, Bingrong Li, Peng Liu, Long Yuan, Kaiyong Zhang, Te Liu e Bimeng Zhang. "Electroacupuncture May Inhibit Oxidative Stress of Premature Ovarian Failure Mice by Regulating Intestinal Microbiota". Oxidative Medicine and Cellular Longevity 2022 (30 de agosto de 2022): 1–13. http://dx.doi.org/10.1155/2022/4362317.

Texto completo da fonte
Resumo:
Premature ovarian failure (POF) is the leading cause of female infertility, and there is no optimal treatment or medication available currently. For POF, electroacupuncture (EA) has been considered a promising therapeutic approach, but the mechanism for this is not clear. In this study, we explored the effects of EA (CV4, ST36, and SP6) on oxidative stress and intestinal microbiota of high-fat and high-sugar- (HFHS-) induced POF mice. The development of mice follicles was observed by hematoxylin and eosin (HE) staining. The serum levels of estrone (E1), estrogen (E2), estriol (E3), and 21-deoxycortisol (21D) were measured by the HPLC-MS/MS method. The concentrations of Fe2+, superoxide dismutase (SOD), hydroxyl radical (·OH), glutathione (GSH), superoxide anion, and malondialdehyde (MDA) were measured by spectrophotometry. The 16S-rDNA sequencing was used to measure many parameters related to the host gut bacteriome and mycobiome composition, relative abundance, and diversity. mRNA expression levels of ferroptosis-related genes were determined by RT-qPCR. After 4 weeks of EA intervention in POF mice, mature follicles were increased and the levels of the sex hormone were improved. SOD activities, antisuperoxide activities, and GSH increased while MDA, ·OH, and Fe2+ decreased. In addition, EA also altered the intestinal microbiota. These results reveal that EA can effectively inhibit ovarian oxidative stress and the accumulation of Fe2+ in POF mice. It may be that the alteration in the intestinal microbiota is one of the potential mechanisms of EA treatment. These findings suggest that EA has clinical potential as a safe treatment for POF.
Estilos ABNT, Harvard, Vancouver, APA, etc.
40

Rolling, Thierry, Bing Zhai, Mergim Gjonbalaj, Nicholas Tosini, Keiko Yasuma-Mitobe, Emily Fontana, Luigi A. Amoretti et al. "Haematopoietic cell transplantation outcomes are linked to intestinal mycobiota dynamics and an expansion of Candida parapsilosis complex species". Nature Microbiology 6, n.º 12 (11 de novembro de 2021): 1505–15. http://dx.doi.org/10.1038/s41564-021-00989-7.

Texto completo da fonte
Estilos ABNT, Harvard, Vancouver, APA, etc.
41

Dinleyici, M., V. Pérez-Brocal, S. Arslanoglu, O. Aydemir, S. Sevuk Ozumut, N. Tekin, Y. Vandenplas, A. Moya e E. C. Dinleyici. "Human milk mycobiota composition: relationship with gestational age, delivery mode, and birth weight". Beneficial Microbes 11, n.º 2 (27 de março de 2020): 151–62. http://dx.doi.org/10.3920/bm2019.0158.

Texto completo da fonte
Resumo:
Intestinal and human milk microbiota studies during infancy have shown variations according to geographical location, delivery mode, gestational age, and mother-related factors during pregnancy. In this study, we performed metagenomic mycobiota analyses of 44 transient and mature human milk among five different groups: mothers of normal spontaneous delivery-term (NS-T), caesarean delivery-term (CS-T), premature (PT), small for gestational age (SGA), and large for gestational age (LGA) infants. Fungi were detected in 80 out of the 88 samples. Regarding the number of observed fungal species, the NS-T group was more homogeneous (less variable) comparing the other groups (P<0.05). In the transient human milk samples, the most abundant species were Saccharomyces cerevisiae (33.3%) and Aspergillus glaucus (27.4%). While A. glaucus (33.7%) was second most abundant species in mature milk, S. cerevisiae disappeared (P<0.01) and Penicillium rubens became the most abundant species (35.5%) (P<0.05). Among the NS-T group, the most abundant species was Malassezia globosa in both transient and mature milk. In contrast, S. cerevisiae was the most abundant species in transient human milk (45.0%) in the CS-T group, but it disappeared in mature milk (P<0.01). In transient milk, M. globosa was only represented 6.0-9.0% of taxa in the PT, SGA, and LGA groups (P<0.05). In transient and mature milk in the PT, SGA and LGA groups, the most abundant species were A. glaucus and P. rubens. In mature milk samples, P. rubens is more abundant in CS-T group, PT group and LGA group, than the NS-T groups (P<0.05 for all). Although fungi constitute only a very small part of the human milk microbiome, we observed some changes that the human milk mycobiota composition varies in caesarean delivery, premature, SGA and LGA groups, comparing the normal spontaneous delivery, as well as differences between transient and mature human milk.
Estilos ABNT, Harvard, Vancouver, APA, etc.
42

Radavelli, Bruna Luísa, Priscila Berti Zanella, Amanda Souza Silva e Valesca Dall’Alba. "Diet, microbiota and inflammatory bowel disease: review". Nutrition & Food Science 48, n.º 2 (12 de março de 2018): 259–71. http://dx.doi.org/10.1108/nfs-07-2017-0156.

Texto completo da fonte
Resumo:
Purpose The purpose of this paper is to verify the possible associations between dietary components and the intestinal microbiota in clinical parameters of inflammatory bowel disease. Design/methodology/approach In this review, a search in PubMed and Bireme databases was performed. The authors included randomized clinical trials published between 2005 and 2017, only in adult humans with Crohn’s disease or ulcerative colitis. Findings Six articles were included by the end of the search. The most widely used intervention was the use of prebiotics, including fructooligosaccharides or fructooligosaccharides with inulin, followed by probiotics. The main findings regarding the microbiota were the increase in the total amount of bacteria and variability (phyla). Clinically, there was improvement in inflammation seen in parameters such as C-reactive protein, interleukins and tumor necrosis factor alpha. Originality/value Dietary interventions, especially from symbiotics, can modulate the microbiota, mainly in relation to time, when compared pre- and post-supplementation, and this positively interferes with clinical parameters of inflammatory bowel diseases. However, the studies were quite heterogeneous in population, methodology, intervention, mycobiota analysis and inflammatory markers.
Estilos ABNT, Harvard, Vancouver, APA, etc.
43

Shin Shin, S., Y. J. Cho, J. Yang, H. K. Kim, P. Rintarhat, M. Park, K. Lagree et al. "P1229 The Role and Niche-Specific Adaptation of Malassezia in patients with Ulcerative colitis". Journal of Crohn's and Colitis 18, Supplement_1 (1 de janeiro de 2024): i2178. http://dx.doi.org/10.1093/ecco-jcc/jjad212.1359.

Texto completo da fonte
Resumo:
Abstract Background Accumulating evidence has underscored the role of gut fungal microbiota (mycobiota) in the development of inflammatory bowel disease. We aimed to isolate a Malassezia strain directly from the human intestine mucosal surface from the patients with ulcerative colitis (UC) and investigated its genome and virulence in comparison with the same fungal species isolated from the human skin. Methods Mucosal lavage samples were collected separately from colonic areas with and without inflammation in patients with UC. Samples from healthy individuals (HT) were obtained in the same manner as from patients with UC at sigmoid or descending colon. Skin samples were taken from HT in our previous work. DNA was extracted from these lavage samples, and fungal isolation was conducted using PCR amplification with ITS4 and ITS5 primers. Comprehensive analysis and comparison of the genomes, transcriptomes, and virulence between M. globosa gut isolates and those of M. globosa strains isolated from the skin were performed. To determine the contribution of M. globosa gut isolates to exacerbating inflammation, 1107 fungal cells were orally gavaged to DSS-induced colitis mouse model for three days. Results Total 56 and 11 intestinal water-lavage samples from 29 UC patients and 11 HT were obtained respectively. The α- and b-diversities of mycobiota showed no significant differences between the groups, patients with UC vs. HT or the sites with inflammation vs. non-inflammation of the patient with UC. Malassezia was the fifth most frequently found fungal genus throughout the samples, and live fungal strains belong to 28 and 7 different species were isolated from the patients with UC and HT, respectively. The patients with UC tend to have higher frequency of M. globosa and M. restricta than HT in their gut mucosal surface with inflammation. Whole genome sequencing showed no specific genomic characteristics between gut-isolated M. globosa and skin-isolated M. globosa. However, gut-isolated M. globosa were suffered more from the higher oxygen levels than the skin isolates in different oxygen concentrations. In a mouse model, gut-isolated M. globosa exhibited a more pronounced exacerbation of DSS-induced colitis and elevated production of inflammatory cytokines, including TNF-a, IL-6, IL-12p40, IL-1b, and IL-18, while the skin isolates showed no difference compared to the negative control (Figure). Conclusion Our data shed new light on the pivotal role of M. globosa in the pathogenesis of UC, highlighting the potential influence of niche-specific adaptations on the virulence of this fungus. These findings provide critical insights into the complex interplay between the member of the gut mycobiota and host health.
Estilos ABNT, Harvard, Vancouver, APA, etc.
44

Prokopyev, V. V., N. V. Kuklina, O. N. Mazko e O. G. Makarova. "Oxidative stress induced by the human microbiota yeast component as a micromycetes pathogenicity factor". Russian Journal of Infection and Immunity 14, n.º 1 (28 de abril de 2024): 175–80. http://dx.doi.org/10.15789/2220-7619-osi-9636.

Texto completo da fonte
Resumo:
The introduction of MALDI-ToF mass spectrometry into routine microbiological diagnostics has led to a widely extended list of identified microorganisms. This phenomenon also affected mycological studies. Rare basidiomycetes and ascomycetes began to be detected in various types of pathological material. It is often difficult to assess a role of isolated micromycetes in the pathogenesis of a certain disease. In addition to generally admitted pathogenicity factors, such as aggression enzymes, adhesive activity, and toxin formation, induction or inhibition of reactive oxygen species can play a prominent role in pathogenesis of infectious diseases. In our study, we evaluated the total prooxidant and total antioxidant activity of cultivated ascomycete and basidiomycete yeasts from the human intestinal mycobiome. The strains of micromycetes assessed here were obtained from a fecal culture study from patients with gastrointestinal tract pathology and healthy people undergoing a routine medical examination. Identification was carried out analyzing morphological, cultural, biochemical properties and confirmed by mass spectrometry. The total prooxidant and antioxidant activity was assessed by the induced or inhibited malondialdehyde formation during Tween-80 oxidation. It was found that the level of total prooxidant activity in Rhodotorula mucilaginosa, Geotrichum candidum, Candida albicans, Pichia kudriavzevii significantly exceeds the level of total antioxidant activity, and the Pichia kudriavzevii prooxidase activity was more than twice as high as in other studied micromycetes. The revealed ability of some fungi to induce oxidase stress can be considered as one of significant pathogenicity factors of microorganisms that cause pathomorphological changes in human intestinal tissues.
Estilos ABNT, Harvard, Vancouver, APA, etc.
45

Dragičević, Paula, Ana Bielen, Jurica Žučko e Sandra Hudina. "The mycobiome of a successful crayfish invader and its changes along the environmental gradient". Animal Microbiome 5, n.º 1 (11 de abril de 2023). http://dx.doi.org/10.1186/s42523-023-00245-9.

Texto completo da fonte
Resumo:
Abstract Background The microbiome plays an important role in biological invasions, since it affects various interactions between host and environment. However, most studies focus on the bacteriome, insufficiently addressing other components of the microbiome such as the mycobiome. Microbial fungi are among the most damaging pathogens in freshwater crayfish populations, colonizing and infecting both native and invasive crayfish species. Invading crayfish may transmit novel fungal species to native populations, but also, dispersal process and characteristics of the novel environment may affect the invaders’ mycobiome composition, directly and indirectly affecting their fitness and invasion success. This study analyzes the mycobiome of a successful invader in Europe, the signal crayfish, using the ITS rRNA amplicon sequencing approach. We explored the mycobiomes of crayfish samples (exoskeletal biofilm, hemolymph, hepatopancreas, intestine), compared them to environmental samples (water, sediment), and examined the differences in fungal diversity and abundance between upstream and downstream segments of the signal crayfish invasion range in the Korana River, Croatia. Results A low number of ASVs (indicating low abundance and/or diversity of fungal taxa) was obtained in hemolymph and hepatopancreas samples. Thus, only exoskeleton, intestine, sediment and water samples were analyzed further. Significant differences were recorded between their mycobiomes, confirming their uniqueness. Generally, environmental mycobiomes showed higher diversity than crayfish-associated mycobiomes. The intestinal mycobiome showed significantly lower richness compared to other mycobiomes. Significant differences in the diversity of sediment and exoskeletal mycobiomes were recorded between different river segments (but not for water and intestinal mycobiomes). Together with the high observed portion of shared ASVs between sediment and exoskeleton, this indicates that the environment (i.e. sediment mycobiome) at least partly shapes the exoskeletal mycobiome of crayfish. Conclusion This study presents the first data on crayfish-associated fungal communities across different tissues, which is valuable given the lack of studies on the crayfish mycobiome. We demonstrate significant differences in the crayfish exoskeletal mycobiome along the invasion range, suggesting that different local environmental conditions may shape the exoskeletal mycobiome during range expansion, while the mycobiome of the internal organ (intestine) remained more stable. Our results provide a basis for assessing how the mycobiome contributes to the overall health of the signal crayfish and its further invasion success.
Estilos ABNT, Harvard, Vancouver, APA, etc.
46

Perlin, Cássio Marques, Larisse Longo, Melina Belén Keingeski, Rafael V. Picon e Mário Reis Álvares-da-Silva. "Gut Mycobiota Changes in Liver Diseases: a Systematic Review". Medical Mycology, 18 de julho de 2023. http://dx.doi.org/10.1093/mmy/myad071.

Texto completo da fonte
Resumo:
Abstract Intestinal fungi play an important role in the health-disease process. We observed that in liver diseases, fungal infections lead to high mortality. In this review we were able to gather and evaluate the available scientific evidence on intestinal mycobiota and liver diseases. We searched PubMed and Embase, using a combination of several entry terms. Only studies in adults ≥ 18 years old with liver disease and published after 2010 were included. We observed that individuals with liver disease have an altered intestinal mycobioma, which accompanies the progression of these diseases. In cirrhotic patients, there are a high number of Candida sp. strains, especially Candida albicans. In early chronic liver disease, there is an increase in alpha diversity at the expense of Candida sp. and conversely, in advanced liver disease, there is a negative correlation between alpha diversity and MELD score. On the other hand, patients with non-alcoholic fatty liver disease demonstrate greater diversity compared to controls. Our study concluded that the evidence on the subject is sparse, with few studies and lack of standardization of outcome measures and reporting, and it was not possible to perform a meta-analysis capable of synthesizing relevant parameters of the human mycobiotic profile. However, certain fungal genera such as Candida play an important role in the context of liver disease and that adults with liver disease have a distinct gut mycobiotic profile from healthy controls.
Estilos ABNT, Harvard, Vancouver, APA, etc.
47

Robinson, Kelsy, Qing Yang, Sydney Stewart, Melanie A. Whitmore e Guolong Zhang. "Biogeography, succession, and origin of the chicken intestinal mycobiome". Microbiome 10, n.º 1 (1 de abril de 2022). http://dx.doi.org/10.1186/s40168-022-01252-9.

Texto completo da fonte
Resumo:
Abstract Background Extensive work has been accomplished to characterize the intestinal bacterial community, known as the microbiota, and its association with host health and disease. However, very little is known about the spatiotemporal development and the origin of a minor intestinal fungal community, known as the mycobiota, in humans and animals, particularly in avian species. Results In this study, we comprehensively characterized the biogeography and succession of the gastrointestinal (GI) mycobiota of broiler chickens and further revealed the fungal sources that are responsible for initial and long-term establishment of the mycobiota in the GI tract. Using Illumina sequencing of the internal transcribed spacer 2 (ITS2) region of fungal rRNA genes, we detected significant spatial and temporal differences in the mycobiota along the GI tract. In contrary to the microbiota, the mycobiota was more diverse in the upper than the lower GI tract with no apparent trend of succession up to 42 days of age. The intestinal mycobiota was dominated by the phyla Ascomycota and Basidiomycota with Gibberella, Aspergillus, and Candida being the most abundant genera. Although the chicken mycobiota was highly dynamic, Fusarium pseudonygamai was dominant throughout the GI tract regardless of age in this study. The core chicken mycobiome consisted of 26 fungal taxa accounting for greater than 85% of the fungal population in each GI location. However, we observed high variations of the intestinal mycobiota among different studies. We also showed that the total fungal population varied greatly from 1.0 × 104 to 1.1 × 106 /g digesta along the GI tract and only accounted for less than 0.06% of the bacteria in day-42 broilers. Finally, we revealed that the mycobiota from the hatchery environment was responsible for initial colonization in the GI tract of newly hatched chickens, but was quickly replaced by the fungi in the diet within 3 days. Conclusions Relative to the intestinal microbiota that consists of trillions of bacteria in hundreds of different species and becomes relatively stabilized as animals age, the chicken intestinal mycobiota is a minor microbial community that is temporally dynamic with limited diversity and no obvious pattern of successive changes. However, similar to the microbiota, the chicken mycobiota is spatially different along the GI tract, although it is more diverse in the upper than the lower GI tract. Dietary fungi are the major source of the intestinal mycobiota in growing chickens.
Estilos ABNT, Harvard, Vancouver, APA, etc.
48

Robinson, Kelsy, Yingping Xiao, Timothy J. Johnson, Binlong Chen, Qing Yang, Wentao Lyu, Jing Wang et al. "Chicken Intestinal Mycobiome: Initial Characterization and Its Response to Bacitracin Methylene Disalicylate". Applied and Environmental Microbiology 86, n.º 13 (1 de maio de 2020). http://dx.doi.org/10.1128/aem.00304-20.

Texto completo da fonte
Resumo:
ABSTRACT The gastrointestinal (GI) tract harbors a diverse population of microorganisms. While much work has been focused on the characterization of the bacterial community, very little is known about the fungal community, or mycobiota, in different animal species and chickens in particular. Here, we characterized the biogeography of the mycobiota along the GI tract of day 28 broiler chicks and further examined its possible shift in response to bacitracin methylene disalicylate (BMD), a commonly used in-feed antibiotic, through Illumina sequencing of the internal transcribed spacer 2 (ITS2) region of fungal rRNA genes. Out of 124 samples sequenced, we identified a total of 468 unique fungal features that belong to four phyla and 125 genera in the GI tract. Ascomycota and Basidiomycota represented 90% to 99% of the intestinal mycobiota, with three genera, i.e., Microascus, Trichosporon, and Aspergillus, accounting for over 80% of the total fungal population in most GI segments. Furthermore, these fungal genera were dominated by Scopulariopsis brevicaulis (Scopulariopsis is the anamorph form of Microascus), Trichosporon asahii, and two Aspergillus species. We also revealed that the mycobiota are more diverse in the upper than lower GI tract. The cecal mycobiota transitioned from being S. brevicaulis dominant on day 14 to T. asahii dominant on day 28. Furthermore, 2-week feeding of 55 mg/kg BMD tended to reduce the cecal mycobiota α-diversity. Taken together, we provided a comprehensive biogeographic view and succession pattern of the chicken intestinal mycobiota and its influence by BMD. A better understanding of intestinal mycobiota may lead to the development of novel strategies to improve animal health and productivity. IMPORTANCE The intestinal microbiota is critical to host physiology, metabolism, and health. However, the fungal community has been often overlooked. Recent studies in humans have highlighted the importance of the mycobiota in obesity and disease, making it imperative that we increase our understanding of the fungal community. The significance of this study is that we revealed the spatial and temporal changes of the mycobiota in the GI tract of the chicken, a nonmammalian species. To our surprise, the chicken intestinal mycobiota is dominated by a limited number of fungal species, in contrast to the presence of hundreds of bacterial taxa in the bacteriome. Additionally, the chicken intestinal fungal community is more diverse in the upper than the lower GI tract, while the bacterial community shows an opposite pattern. Collectively, this study lays an important foundation for future work on the chicken intestinal mycobiome and its possible manipulation to enhance animal performance and disease resistance.
Estilos ABNT, Harvard, Vancouver, APA, etc.
49

James, Steve, Aimee Parker, Catherine Purse, Dave Baker, Andrea Telatin, Simon Funnell e Simon Carding. "A Fungal Foray in an NHP Gut Microbiome". Access Microbiology 4, n.º 7 (1 de junho de 2022). http://dx.doi.org/10.1099/acmi.byg2021.po0004.

Texto completo da fonte
Resumo:
The cynomolgus macaque, Macaca fascicularis, is a non-human primate (NHP) widely used in biomedical research as it shares behavioural, genetic, immunological and physiological similarities with humans. These similarities may extend to the enteric microbiome, with some microbial taxa common to both humans and NHPs. However, to date, the majority of these microbial surveys have focused on the prokaryome, and have largely ignored or overlooked the NHP gut mycobiome. To address this shortfall, we have undertaken a region-by-region taxonomic survey of the cynomolgus intestinal mycobiota, from duodenum to distal colon, of ten captive animals of differing age. Using a high-throughput ITS1 amplicon sequencing-based approach, we found that fungi from the Ascomycota phylum dominate the cynomolgus enteric mycobiota. The budding yeast genus Kazachstania was most abundant, with K. pintolopesii and K. telluris highly prevalent, and the predominant species in many of the intestinal samples. However, while K. pintolopesii was present throughout the primate GI tract, K. telluris was found mainly in the small intestine. In this study, K. pintolopesii was identified as the dominant enteric fungus in captive cynomolgus macaques. This contrasts with humans, where Candida albicans is a common member of the intestinal microbiota. To our knowledge, this is the first time K. pintolopesii has been identified as a primate gut commensal.
Estilos ABNT, Harvard, Vancouver, APA, etc.
50

Moreno-Sabater, Alicia, Gaelle Autaa, Delphine Sterlin, Amenie Jerbi, Remy Villette, Johanna B. Holm, Christophe Parizot et al. "Systemic anti-commensal response to fungi analyzed by flow cytometry is related to gut mycobiome ecology". Microbiome 8, n.º 1 (15 de novembro de 2020). http://dx.doi.org/10.1186/s40168-020-00924-8.

Texto completo da fonte
Resumo:
Abstract Background Interest for the study of gut mycobiota in relation with human health and immune homeostasis has increased in the last years. From this perspective, new tools to study the immune/fungal interface are warranted. Systemic humoral immune responses could reflect the dynamic relationships between gut mycobiota and immunity. Using a novel flow cytometry technology (Fungi-Flow) to determine immunoglobulin (Ig) responses to fungi, we studied the relationships between gut mycobiota and systemic humoral anti-commensal immunity. Results The Fungi-Flow method allows a sensitive and specific measurement of systemic IgG responses against 17 commensal and environmental fungi from the two main divisions; Ascomycota and Basidiomycota. IgG responses exhibited a high inter-individual variability. Anti-commensal IgG responses were contrasted with the relative abundance, alpha-diversity, and intra-genus richness of fungal species in gut mycobiota of twenty healthy donors. Categorization of gut mycobiota composition revealed two differentiated fungal ecosystems. Significant difference of anti-Saccharomyces systemic IgG responses were observed in healthy donors stratified according to the fungal ecosystem colonizing their gut. A positive and significant correlation was observed between the variety of IgG responses against fungal commensals and intestinal alpha-diversity. At the level of intra-genus species richness, intense IgG responses were associated with a low intra-genus richness for known pathobionts, but not commensals. Conclusions Fungi-Flow allows an easy and reliable measure of personalized humoral responses against commensal fungi. Combining sequencing technology with our novel Fungi-Flow immunological method, we propose that there are at least two defined ecosystems in the human gut mycobiome associated with systemic humoral responses. Fungi-Flow opens new opportunities to improve our knowledge about the impact of mycobiota in humoral anti-commensal immunity and homeostasis.
Estilos ABNT, Harvard, Vancouver, APA, etc.
Oferecemos descontos em todos os planos premium para autores cujas obras estão incluídas em seleções literárias temáticas. Contate-nos para obter um código promocional único!

Vá para a bibliografia