Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.786
Filter
1.
PLoS One ; 19(7): e0307079, 2024.
Article in English | MEDLINE | ID: mdl-39012882

ABSTRACT

BACKGROUND: Most of the current bacteriophages (phages) are mostly isolated from environments. However, phages isolated from feces might be more specific to the bacteria that are harmful to the host. Meanwhile, some phages from the environment might affect non-pathogenic bacteria for the host. METHODS: Here, bacteriophages isolated from mouse feces were intratracheally (IT) or intravenously (IV) administered in pneumonia mice caused by Pseudomonas aeruginosa at 2 hours post-intratracheal bacterial administration. As such, the mice with phage treatment, using either IT or IV administration, demonstrated less severe pneumonia as indicated by mortality, serum cytokines, bacteremia, bacterial abundance in bronchoalveolar lavage fluid (BALF), and neutrophil extracellular traps (NETs) in lung tissue (immunofluorescence of neutrophil elastase and myeloperoxidase). RESULTS: Interestingly, the abundance of phages in BALF from the IT and IV injections was similar, supporting a flexible route of phage administration. With the incubation of bacteria with neutrophils, the presence of bacteriophages significantly improved bactericidal activity, but not NETs formation, with the elevated supernatant IL-6 and TNF-α, but not IL-1ß. In conclusion, our findings suggest that bacteriophages against Pseudomonas aeruginosa can be discovered from feces of the host. CONCLUSIONS: The phages attenuate pneumonia partly through an enhanced neutrophil bactericidal activity, but not via inducing NETs formation. The isolation of phages from the infected hosts themselves might be practically useful for future treatment. More studies are warranted.


Subject(s)
Feces , Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Pseudomonas aeruginosa/virology , Feces/microbiology , Feces/virology , Mice , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Bronchoalveolar Lavage Fluid/microbiology , Bronchoalveolar Lavage Fluid/virology , Neutrophils/immunology , Bacteriophages/isolation & purification , Bacteriophages/physiology , Extracellular Traps , Pneumonia/microbiology , Pneumonia/therapy , Pneumonia/virology , Cytokines/metabolism , Cytokines/blood , Phage Therapy/methods , Female , Lung/microbiology , Lung/virology , Pneumonia, Bacterial/therapy , Pneumonia, Bacterial/microbiology
2.
Curr Microbiol ; 81(9): 267, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39003673

ABSTRACT

In this study, we evaluated the impact of human gut microbiota on the immune pathways in the respiratory tract using a gnotobiotic (Gn) piglet model. We humanized piglets with rural and urban infant fecal microbiota (RIFM and UIFM, respectively) and then infected them with a H1N1 swine influenza virus. We analyzed the microbial diversity and structure of the intestinal and respiratory tracts of the piglets before and after the influenza virus infection and measured the viral load and immune responses. We found that the viral load in the upper respiratory tract of UIFM transplanted piglets was higher than their rural cohorts (RIFM), while virus-specific antibody responses were comparable. The relative cytokine gene expression in the tracheobronchial (respiratory tract) and mesenteric (gastrointestinal) lymph nodes, lungs, blood, and spleen of RIFM and UIFM piglets revealed a trend in reciprocal regulation of proinflammatory, innate, and adaptive immune-associated cytokines as well as the frequency of T-helper/memory cells, cytotoxic T cells, and myeloid immune cell subsets. We also observed different phylum-level shifts of the fecal microbiota in response to influenza virus infection between the two piglet groups, suggesting the potential impact of the gut microbiota on the immune responses to influenza virus infection and lung microbiota. In conclusion, Gn piglets humanized with diverse infant fecal microbiota had differential immune regulation, with UIFM favoring the activation of proinflammatory immune mediators following an influenza virus infection compared to their rural RIFM cohorts. Furthermore, Gn piglets can be a useful model in investigating the impact of diverse human microbiota of the gastrointestinal tract, probably also the respiratory tract, on respiratory health and testing specific probiotic- or prebiotic-based therapeutics.


Subject(s)
Cytokines , Disease Models, Animal , Feces , Gastrointestinal Microbiome , Germ-Free Life , Immunity, Mucosal , Influenza A Virus, H1N1 Subtype , Animals , Swine , Feces/microbiology , Feces/virology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Cytokines/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Viral Load , Infant , Influenza, Human/immunology , Influenza, Human/microbiology , Influenza, Human/virology
3.
J Med Virol ; 96(7): e29809, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39016466

ABSTRACT

Pancreatic cancer (PC) is a highly aggressive malignancy with a poor prognosis, making early diagnosis crucial for improving patient outcomes. While the gut microbiome, including bacteria and viruses, is believed to be essential in cancer pathogenicity, the potential contribution of the gut virome to PC remains largely unexplored. In this study, we conducted a comparative analysis of the gut viral compositional and functional profiles between PC patients and healthy controls, based on fecal metagenomes from two publicly available data sets comprising a total of 101 patients and 82 healthy controls. Our results revealed a decreasing trend in the gut virome diversity of PC patients with disease severity. We identified significant alterations in the overall viral structure of PC patients, with a meta-analysis revealing 219 viral operational taxonomic units (vOTUs) showing significant differences in relative abundance between patients and healthy controls. Among these, 65 vOTUs were enriched in PC patients, and 154 were reduced. Host prediction revealed that PC-enriched vOTUs preferentially infected bacterial members of Veillonellaceae, Enterobacteriaceae, Fusobacteriaceae, and Streptococcaceae, while PC-reduced vOTUs were more likely to infect Ruminococcaceae, Lachnospiraceae, Clostridiaceae, Oscillospiraceae, and Peptostreptococcaceae. Furthermore, we constructed random forest models based on the PC-associated vOTUs, achieving an optimal average area under the curve (AUC) of up to 0.879 for distinguishing patients from controls. Through additional 10 public cohorts, we demonstrated the reproducibility and high specificity of these viral signatures. Our study suggests that the gut virome may play a role in PC development and could serve as a promising target for PC diagnosis and therapeutic intervention. Future studies should further explore the underlying mechanisms of gut virus-bacteria interactions and validate the diagnostic models in larger and more diverse populations.


Subject(s)
Feces , Gastrointestinal Microbiome , Metagenomics , Pancreatic Neoplasms , Virome , Humans , Pancreatic Neoplasms/virology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/microbiology , Gastrointestinal Microbiome/genetics , Metagenomics/methods , Feces/virology , Feces/microbiology , Viruses/isolation & purification , Viruses/genetics , Viruses/classification , Metagenome , Bacteria/isolation & purification , Bacteria/classification , Bacteria/genetics , Middle Aged , Male , Female , Aged , Case-Control Studies
4.
Genome Biol ; 25(1): 177, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965579

ABSTRACT

Identifying viruses from metagenomes is a common step to explore the virus composition in the human gut. Here, we introduce VirRep, a hybrid language representation learning framework, for identifying viruses from human gut metagenomes. VirRep combines a context-aware encoder and an evolution-aware encoder to improve sequence representation by incorporating k-mer patterns and sequence homologies. Benchmarking on both simulated and real datasets with varying viral proportions demonstrates that VirRep outperforms state-of-the-art methods. When applied to fecal metagenomes from a colorectal cancer cohort, VirRep identifies 39 high-quality viral species associated with the disease, many of which cannot be detected by existing methods.


Subject(s)
Gastrointestinal Microbiome , Metagenome , Humans , Viruses/genetics , Feces/virology , Metagenomics/methods , Software , Colorectal Neoplasms/virology , Colorectal Neoplasms/genetics
5.
Virol J ; 21(1): 153, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38972989

ABSTRACT

Wild waterfowl serve as a reservoir of some astroviruses. Fecal samples from wild waterfowl collected at Hong Kong's Marshes were tested using pan-astrovirus reverse transcription-PCR. Positive samples underwent subsequent host identification using DNA barcoding. Based on deduced partial sequences, noteworthy samples from three astrovirus groups (mammalian, avian and unclassified astroviruses) were further analyzed by next-generation sequencing. One sample of Avastrovirus 4 clade, MP22-196, had a nearly complete genome identified. The results of ORF2 phylogenetic analysis and genetic distance analysis indicate that Avastrovirus 4 is classified as a distinct subclade within Avastrovirus. MP22-196 has typical astrovirus genome characteristics. The unique characteristics and potential differences of this genome, compared to other avian astrovirus sequences, involve the identification of a modified sgRNA sequence situated near the ORF2 start codon, which precedes the ORF1b stop codon. Additionally, the 3' UTR of MP22-196 is shorter than other avian astroviruses. This study expands our understanding of the Avastrovirus 4 clade.


Subject(s)
Astroviridae Infections , Birds , Feces , Genetic Variation , Genome, Viral , Phylogeny , Animals , Hong Kong , Birds/virology , Feces/virology , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Animals, Wild/virology , Bird Diseases/virology , High-Throughput Nucleotide Sequencing , Avastrovirus/genetics , Avastrovirus/classification , Avastrovirus/isolation & purification , RNA, Viral/genetics , Open Reading Frames , Astroviridae/genetics , Astroviridae/isolation & purification , Astroviridae/classification
6.
PLoS Negl Trop Dis ; 18(7): e0012228, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38976836

ABSTRACT

BACKGROUND: Viral gastrointestinal infections remain a major public health concern in developing countries. In Burkina Faso, there are very limited updated data on the circulating viruses and their genetic diversity. OBJECTIVES: This study investigates the detection rates and characteristics of rotavirus A (RVA), norovirus (NoV), sapovirus (SaV) and human astrovirus (HAstV) in patients of all ages with acute gastrointestinal infection in urban and rural areas. STUDY DESIGN & METHODS: From 2018 to 2021, stool samples from 1,295 patients with acute gastroenteritis were collected and screened for RVA, NoV, SaV and HAstV. Genotyping and phylogenetic analyses were performed on a subset of samples. RESULTS: At least one virus was detected in 34.1% of samples. NoV and SaV were predominant with detection rates of respectively 10.5 and 8.8%. We identified rare genotypes of NoV GII, RVA and HAstV, recombinant HAstV strains and a potential zoonotic RVA transmission event. CONCLUSIONS: We give an up-to-date epidemiological picture of enteric viruses in Burkina Faso, showing a decrease in prevalence but a high diversity of circulating strains. However, viral gastroenteritis remains a public health burden, particularly in pediatric settings. Our data advocate for the implementation of routine viral surveillance and updated management algorithms for diarrheal disease.


Subject(s)
Gastroenteritis , Genetic Variation , Genotype , Norovirus , Phylogeny , Rotavirus , Rural Population , Humans , Burkina Faso/epidemiology , Gastroenteritis/virology , Gastroenteritis/epidemiology , Child, Preschool , Infant , Child , Male , Female , Rotavirus/genetics , Rotavirus/classification , Rotavirus/isolation & purification , Adolescent , Adult , Norovirus/genetics , Norovirus/classification , Norovirus/isolation & purification , Young Adult , Feces/virology , Sapovirus/genetics , Sapovirus/isolation & purification , Sapovirus/classification , Middle Aged , Urban Population , Infant, Newborn , Caliciviridae Infections/epidemiology , Caliciviridae Infections/virology , Mamastrovirus/genetics , Mamastrovirus/classification , Mamastrovirus/isolation & purification , Aged , Prevalence
7.
Microbiome ; 12(1): 122, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38970126

ABSTRACT

BACKGROUND: Fecal microbiota transplantation (FMT) is a therapeutic intervention used to treat diseases associated with the gut microbiome. In the human gut microbiome, phages have been implicated in influencing human health, with successful engraftment of donor phages correlated with FMT treatment efficacy. The impact that gastrointestinal phages exert on human health has primarily been connected to their ability to modulate the bacterial communities in the gut. Nonetheless, how FMT affects recipients' phage populations, and in turn, how this influences the gut environment, is not yet fully understood. In this study, we investigated the effects of FMT on the phageome composition of participants within the Gut Bugs Trial (GBT), a double-blind, randomized, placebo-controlled trial that investigated the efficacy of FMT in treating obesity and comorbidities in adolescents. Stool samples collected from donors at the time of treatment and recipients at four time points (i.e., baseline and 6 weeks, 12 weeks, and 26 weeks post-intervention), underwent shotgun metagenomic sequencing. Phage sequences were identified and characterized in silico to examine evidence of phage engraftment and to assess the extent of FMT-induced alterations in the recipients' phageome composition. RESULTS: Donor phages engrafted stably in recipients following FMT, composing a significant proportion of their phageome for the entire course of the study (33.8 ± 1.2% in females and 33.9 ± 3.7% in males). Phage engraftment varied between donors and donor engraftment efficacy was positively correlated with their phageome alpha diversity. FMT caused a shift in recipients' phageome toward the donors' composition and increased phageome alpha diversity and variability over time. CONCLUSIONS: FMT significantly altered recipients' phage and, overall, microbial populations. The increase in microbial diversity and variability is consistent with a shift in microbial population dynamics. This proposes that phages play a critical role in modulating the gut environment and suggests novel approaches to understanding the efficacy of FMT in altering the recipient's microbiome. TRIAL REGISTRATION: The Gut Bugs Trial was registered with the Australian New Zealand Clinical Trials Registry (ACTR N12615001351505). Trial protocol: the trial protocol is available at https://bmjopen.bmj.com/content/9/4/e026174 . Video Abstract.


Subject(s)
Bacteriophages , Fecal Microbiota Transplantation , Feces , Gastrointestinal Microbiome , Obesity , Humans , Fecal Microbiota Transplantation/methods , Bacteriophages/physiology , Bacteriophages/classification , Bacteriophages/isolation & purification , Bacteriophages/genetics , Feces/microbiology , Feces/virology , Obesity/therapy , Obesity/microbiology , Double-Blind Method , Female , Adolescent , Male , Bacteria/classification , Bacteria/virology , Bacteria/genetics , Metagenomics/methods , Treatment Outcome
8.
Vopr Virusol ; 69(3): 255-265, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38996374

ABSTRACT

INTRODUCTION: Bats are natural reservoirs of coronaviruses (Coronaviridae), which have caused three outbreaks of human disease SARS, MERS and COVID-19 or SARS-2 over the past decade. The purpose of the work is to study the diversity of coronaviruses among bats inhabiting the foothills and mountainous areas of the Republics of Dagestan, Altai and the Kemerovo region. MATERIALS AND METHODS: Samples of bat oral swabs and feces were tested for the presence of coronavirus RNA by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: It has been shown that the greater horseshoe bats (Rhinolophus ferrumequinum), inhabiting the Republic of Dagestan, are carriers of two different coronaviruses. One of the two coronaviruses is a member of the Sarbecovius subgenus of the Betacoronavirus genus, which includes the causative agents of SARS and COVID-19. The second coronavirus is assigned to the Decacovirus subgenus of the Alphacoronavirus genus and is most similar to viruses identified among Rhinolophus spp. from European and Middle Eastern countries. In the Altai Republic and Kemerovo region, coronaviruses belonging to the genus Alphacoronavirus, subgenus Pedacovirus, were found in the smooth-nosed bats: Ikonnikov`s bat (Myotis ikonnikovi) and the eastern bat (Myotis petax). The virus from the Altai Republic from M. ikonnikovi is close to viruses from Japan and Korea, as well as viruses from Myotis spp. from European countries. The virus from the Kemerovo region from M. petax groups with coronaviruses from Myotis spp. from Asian countries and is significantly different from coronaviruses previously discovered in the same natural host.


Subject(s)
Chiroptera , Animals , Chiroptera/virology , Siberia/epidemiology , Phylogeny , Disease Reservoirs/virology , Coronavirus/genetics , Coronavirus/isolation & purification , Coronavirus/classification , Humans , Feces/virology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , COVID-19/virology , COVID-19/epidemiology , COVID-19/veterinary , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Coronavirus Infections/epidemiology
9.
Vet Res ; 55(1): 87, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982477

ABSTRACT

Hepatitis E virus (HEV) is a major cause of viral hepatitis worldwide. Pigs are the natural host of HEV genotype 3 and the main reservoir of HEV. As the host range of HEV genotype 3 expands, the possibility that HEV from various species can be transmitted to humans via pigs is increasing. We investigated the potential cross-species transmission of HEV by infecting minipigs with swine HEV (swHEV), rabbit HEV (rbHEV), and human HEV (huHEV) and examining their histopathological characteristics and distribution in various organs. Fifteen specific-pathogen-free Yucatan minipigs were infected with swHEV, rbHEV, huHEV, or a mock control. In the present study, we analysed faecal shedding, viremia, and serological parameters over a seven-week period. Our results indicated that swHEV exhibited more robust shedding and viremia than non-swHEVs. Only swHEV affected the serological parameters, suggesting strain-specific differences. Histopathological examination revealed distinct patterns in the liver, pancreas, intestine, and lymphoid tissues after infection with each HEV strain. Notably, all three HEVs induced histopathological changes in the pancreas, supporting the association of HEVs with acute pancreatitis. Our results also identified skeletal muscle as a site of HEV antigen presence, suggesting a potential link to myositis. In conclusion, this study provides valuable insights into the infection dynamics of different HEV strains in minipigs, emphasizing the strain-specific variations in virological, serological, and histological parameters. The observed differences in infection kinetics and tissue tropism will contribute to our understanding of HEV pathogenesis and the potential for cross-species transmission.


Subject(s)
Hepatitis E virus , Hepatitis E , Swine Diseases , Swine, Miniature , Animals , Swine , Hepatitis E/veterinary , Hepatitis E/virology , Hepatitis E/transmission , Hepatitis E virus/physiology , Swine Diseases/virology , Swine Diseases/transmission , Swine Diseases/pathology , Specific Pathogen-Free Organisms , Rabbits , Virus Shedding , Humans , Feces/virology , Female , Viremia/veterinary , Viremia/virology
10.
BMC Vet Res ; 20(1): 305, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982473

ABSTRACT

BACKGROUND: Pigeon Rotavirus A (RVA) infection has been confirmed in pigeons in the last decade as a cause of Young Pigeon Disease (YPD). Although YPD has been known for many years to date, no studies have been conducted to track the spread of RVA infection in pigeons during the racing season. The presented research aims to determine the course of RVA infection during the flights of young racing pigeons in the summer season, in one of the districts in the Mazovian Voivodeship in Poland. RESULTS: Faecal samples of pigeons collected from transport baskets in vehicles transporting pigeons to the starting point were tested. The quantitative RT-PCR (qRT-PCR) was used to detect the genetic material of RVA. Samples taken during 6 flights were analysed. The study showed a percentage increase in infections up to the fourth flight of pigeons, and then their decrease. With Cq values below 20, breeders did not participate in the next flight and/or reported disease in the flock. With positive Cq values of 20 to 30, clinical signs of disease were not reported. Of the 76 breeders participating in the races, at least one positive result was found in 46 (60.5%). Including the occurrence of the disease during the racing season was reported by 11 breeders (14.4%). The main clinical signs in sick pigeons were vomiting, diarrhea and stowed crop. The tested pigeons were not vaccinated against RVA. CONCLUSIONS: During training and racing of pigeons, it is not possible to avoid exposing them to pathogens, including RVA, regardless of whether pigeons from different breeders are placed in the same baskets or are in separate baskets. However, after four flights the number of new cases of the disease decreases which indicates the development of immunity. The qRT-PCR test is useful in the diagnosis and differentiation of clinical (Cq below 20) and subclinical RVA infections in racing pigeons.


Subject(s)
Bird Diseases , Columbidae , Feces , Rotavirus Infections , Rotavirus , Seasons , Animals , Columbidae/virology , Rotavirus Infections/veterinary , Rotavirus Infections/virology , Rotavirus Infections/epidemiology , Bird Diseases/virology , Bird Diseases/epidemiology , Rotavirus/isolation & purification , Feces/virology , Poland/epidemiology
11.
J Med Virol ; 96(7): e29777, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38949212

ABSTRACT

Hepatitis E virus (HEV) is a prevalent pathogen responsible for acute viral hepatitis, HEV genotypes 3 and 4 infections causing zoonotic infections. Currently, the nucleotide similarity analysis between humans and pigs for HEV genotype 4 is limited. In this study, stool samples from an HEV-infected patient who is a pig farmer and from pigs were collected to obtain the near full-length genome of HEV, phylogenetic trees were constructed for genotyping, and similarity of HEV sequences was analyzed. The results showed that HEV-RNA was detected in the stool samples from the patient and six pigs (6/30, 20.0%). Both HEV subtype in the patient and pigs was 4b. Additionally, similarity analysis showed that the range was 99.875%-99.944% between the patient and pigs at the nucleotide level. Four isolates of amino acid sequences (ORFs 1-3) from pigs were 100% identical to the patient. Phylogenetic tree and similarity analysis of an additional nine HEV sequences isolated from other patients in this region showed that the HEV sequence from the pig farmer had the closest relationship with the pigs from his farm rather than other sources of infection in this region. This study provides indirect evidences for HEV subtype 4b can be transmitted from pigs to humans at the nucleotide level. Further research is needed to explore the characteristics of different HEV subtypes.


Subject(s)
Feces , Genome, Viral , Genotype , Hepatitis E virus , Hepatitis E , Phylogeny , RNA, Viral , Swine Diseases , Animals , Hepatitis E virus/genetics , Hepatitis E virus/classification , Hepatitis E virus/isolation & purification , Swine , Hepatitis E/virology , Hepatitis E/veterinary , Hepatitis E/epidemiology , China/epidemiology , Humans , Feces/virology , Swine Diseases/virology , RNA, Viral/genetics , Male , Sequence Analysis, DNA
12.
BMC Microbiol ; 24(1): 264, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026166

ABSTRACT

BACKGROUND: More than 90% of colorectal cancer (CRC) arises from advanced adenomas (AA) and gut microbes are closely associated with the initiation and progression of both AA and CRC. OBJECTIVE: To analyze the characteristic microbes in AA. METHODS: Fecal samples were collected from 92 AA and 184 negative control (NC). Illumina HiSeq X sequencing platform was used for high-throughput sequencing of microbial populations. The sequencing results were annotated and compared with NCBI RefSeq database to find the microbial characteristics of AA. R-vegan package was used to analyze α diversity and ß diversity. α diversity included box diagram, and ß diversity included Principal Component Analysis (PCA), principal co-ordinates analysis (PCoA), and non-metric multidimensional scaling (NMDS). The AA risk prediction models were constructed based on six kinds of machine learning algorithms. In addition, unsupervised clustering methods were used to classify bacteria and viruses. Finally, the characteristics of bacteria and viruses in different subtypes were analyzed. RESULTS: The abundance of Prevotella sp900557255, Alistipes putredinis, and Megamonas funiformis were higher in AA, while the abundance of Lilyvirus, Felixounavirus, and Drulisvirus were also higher in AA. The Catboost based model for predicting the risk of AA has the highest accuracy (bacteria test set: 87.27%; virus test set: 83.33%). In addition, 4 subtypes (B1V1, B1V2, B2V1, and B2V2) were distinguished based on the abundance of gut bacteria and enteroviruses (EVs). Escherichia coli D, Prevotella sp900557255, CAG-180 sp000432435, Phocaeicola plebeiuA, Teseptimavirus, Svunavirus, Felixounavirus, and Jiaodavirus are the characteristic bacteria and viruses of 4 subtypes. The results of Catboost model indicated that the accuracy of prediction improved after incorporating subtypes. The accuracy of discovery sets was 100%, 96.34%, 100%, and 98.46% in 4 subtypes, respectively. CONCLUSION: Prevotella sp900557255 and Felixounavirus have high value in early warning of AA. As promising non-invasive biomarkers, gut microbes can become potential diagnostic targets for AA, and the accuracy of predicting AA can be improved by typing.


Subject(s)
Adenoma , Bacteria , Colorectal Neoplasms , Feces , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/genetics , Bacteria/genetics , Bacteria/classification , Bacteria/isolation & purification , Adenoma/microbiology , Adenoma/virology , Feces/microbiology , Feces/virology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/virology , Male , Middle Aged , Female , Viruses/isolation & purification , Viruses/classification , Viruses/genetics , Viruses/pathogenicity , High-Throughput Nucleotide Sequencing , Aged , Machine Learning
13.
Microbiome ; 12(1): 130, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026313

ABSTRACT

BACKGROUND: The gut virome has been implicated in inflammatory bowel disease (IBD), yet a full understanding of the gut virome in IBD patients, especially across diverse geographic populations, is lacking. RESULTS: In this study, we conducted a comprehensive gut virome-wide association study in a Chinese cohort of 71 IBD patients (15 with Crohn's disease and 56 with ulcerative colitis) and 77 healthy controls via viral-like particle (VLP) and bulk virome sequencing of their feces. By utilizing an integrated gut virus catalog tailored to the IBD virome, we revealed fundamental alterations in the gut virome in IBD patients. These characterized 139 differentially abundant viral signatures, including elevated phages predicted to infect Escherichia, Klebsiella, Enterococcus_B, Streptococcus, and Veillonella species, as well as IBD-depleted phages targeting Prevotella, Ruminococcus_E, Bifidobacterium, and Blautia species. Remarkably, these viral signatures demonstrated high consistency across diverse populations such as those in Europe and the USA, emphasizing their significance and broad relevance in the disease context. Furthermore, fecal virome transplantation experiments verified that the colonization of these IBD-characterized viruses can modulate experimental colitis in mouse models. CONCLUSIONS: Building upon these insights into the IBD gut virome, we identified potential biomarkers for prognosis and therapy in IBD patients, laying the foundation for further exploration of viromes in related conditions. Video Abstract.


Subject(s)
Feces , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Virome , Humans , Gastrointestinal Microbiome/genetics , Animals , Feces/virology , Feces/microbiology , Mice , Inflammatory Bowel Diseases/virology , Inflammatory Bowel Diseases/microbiology , Female , Male , Adult , Middle Aged , Crohn Disease/virology , Crohn Disease/microbiology , Bacteriophages/genetics , Bacteriophages/isolation & purification , Colitis, Ulcerative/virology , Colitis, Ulcerative/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , China , Fecal Microbiota Transplantation , Case-Control Studies , Viruses/classification , Viruses/isolation & purification , Viruses/genetics
14.
Microbiome ; 12(1): 119, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951925

ABSTRACT

BACKGROUND: Fecal microbiota transplantation (FMT) and fecal virome transplantation (FVT, sterile filtrated donor feces) have been effective in treating recurrent Clostridioides difficile infections, possibly through bacteriophage-mediated modulation of the gut microbiome. However, challenges like donor variability, costly screening, coupled with concerns over pathogen transfer (incl. eukaryotic viruses) with FMT or FVT hinder their wider clinical application in treating less acute diseases. METHODS: To overcome these challenges, we developed methods to broaden FVT's clinical application while maintaining efficacy and increasing safety. Specifically, we employed the following approaches: (1) chemostat-fermentation to reproduce the bacteriophage FVT donor component and remove eukaryotic viruses (FVT-ChP), (2) solvent-detergent treatment to inactivate enveloped viruses (FVT-SDT), and (3) pyronin-Y treatment to inhibit RNA virus replication (FVT-PyT). We assessed the efficacy of these processed FVTs in a C. difficile infection mouse model and compared them with untreated FVT (FVT-UnT), FMT, and saline. RESULTS: FVT-SDT, FVT-UnT, and FVT-ChP reduced the incidence of mice reaching the humane endpoint (0/8, 2/7, and 3/8, respectively) compared to FMT, FVT-PyT, and saline (5/8, 7/8, and 5/7, respectively) and significantly reduced the load of colonizing C. difficile cells and associated toxin A/B levels. There was a potential elimination of C. difficile colonization, with seven out of eight mice treated with FVT-SDT testing negative with qPCR. In contrast, all other treatments exhibited the continued presence of C. difficile. Moreover, the results were supported by changes in the gut microbiome profiles, cecal cytokine levels, and histopathological findings. Assessment of viral engraftment following FMT/FVT treatment and host-phage correlations analysis suggested that transfer of phages likely were an important contributing factor associated with treatment efficacy. CONCLUSIONS: This proof-of-concept study shows that specific modifications of FVT hold promise in addressing challenges related to donor variability and infection risks. Two strategies lead to treatments significantly limiting C. difficile colonization in mice, with solvent/detergent treatment and chemostat propagation of donor phages emerging as promising approaches. Video Abstract.


Subject(s)
Bacteriophages , Clostridioides difficile , Clostridium Infections , Fecal Microbiota Transplantation , Feces , Gastrointestinal Microbiome , Fecal Microbiota Transplantation/methods , Animals , Mice , Bacteriophages/physiology , Bacteriophages/isolation & purification , Clostridium Infections/therapy , Clostridium Infections/microbiology , Feces/microbiology , Feces/virology , Disease Models, Animal , Humans , Mice, Inbred C57BL , Female
15.
Zhonghua Er Ke Za Zhi ; 62(7): 643-648, 2024 Jul 02.
Article in Chinese | MEDLINE | ID: mdl-38955682

ABSTRACT

Objective: To investigate the association between intestinal colonization of segmented filamentous bacteria (SFB) and the risk of rotavirus infection, and the possible mechanisms by which SFB resist rotavirus infection. Methods: This case-control study enrolled 50 children aged 0 to 5 years who present to the outpatient Department of Children's Hospital, Zhejiang University School of Medicine with diarrhea and positive stool tests for rotavirus. The children were divided into rotavirus enteritis group and control group consisting of 55 children with non-gastrointestinal and non-infectious surgical diseases.The age and sex composition of the two groups was matched. The DNA of the fecal flora was extracted and SFB was detected by real-time fluorescence quantitative PCR analysis. The children in the rotavirus enteritis group and the control group were subgrouped by age and sex to analyze the differences in SFB positivity rates between different groups, and further compare and analyze the differences in SFB positivity rates between these two groups of children in the ≤2 years old subgroup and the >2-5 years old subgroup. Neutralization test was performed with p3340 protein and rotavirus to determine the relationship between rotavirus infection rate and p3340 concentration in Vero cells. χ2 test or Fisher's exact probability method was used for comparison between the two groups. Results: There were 50 children in the rotavirus enteritis group with an age of (1.7±0.9) years, and 55 children in the control group with an age of (1.8±1.1) years. The positive rate of SFB in children with rotavirus enteritis showed a declining trend across ages groups, with the highest rate of 10/14 in the ≤1 year old group, followed by 67% (14/21) in the >1-2 years old group, 9/15 in the >2-5 years old group, and there was no statistically significant difference (P=0.867). The positive rate of SFB in the control group was 12/15 in the ≤1 year old group, 95% (19/20) in the >1-2 years old group, 50% (10/20) in the >2-5 years old group, with statistical significance (P=0.004). The positive rate of SFB in children with rotavirus enteritis was 74% (20/27) in males and 56% (13/23) in females (χ2=1.71, P=0.192). In the control group, it was 79% (22/28) in males and 70% (19/27) in females (χ2=0.49, P=0.485). The positive rate of SFB was 66% (33/50) in the rotavirus enteritis group and 75% (41/55) in the control group, with no statistically significant (χ2=0.56, P=0.454). In the children ≤2 years old, the SFB positivity rate was 69% (24/35) in the rotavirus enteritis group and 89% (31/35) in the control group, with a statistically significant difference (χ2=4.16, P=0.041). However, in the children >2-5 years old, no statistically significant difference was observed, with the positive rate of SFB being 9/15 in the rotavirus enteritis group and 50% (10/20) in the control group (P=0.734). Pearson correlation analysis revealed a negative correlation between rotavirus infection and SFB positivity (r=-0.87,P<0.001). As the concentration of the p3340 specific protein increased, the luminescence intensity of the luciferase in the Vero cells, which were suitable for cultivating rotavirus, exhibited a decreasing trend (F=4.17, P=0.001). Conclusions: SFB colonization in infants less than 2 years old is associated with a reduced risk of rotavirus infection. Cloning of specific SFB functional protein p3340 neutralizes rotavirus infection of Vero cells, and this mechanism of targeting rotavirus infection differs from the common antiviral mechanism.


Subject(s)
Feces , Rotavirus Infections , Rotavirus , Humans , Infant , Male , Female , Case-Control Studies , Child, Preschool , Feces/virology , Feces/microbiology , Diarrhea/virology , Diarrhea/microbiology , Enteritis/virology , Enteritis/microbiology , Infant, Newborn , Intestines/virology , Intestines/microbiology , Animals
16.
J Med Virol ; 96(7): e29781, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961767

ABSTRACT

Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is a serious and common extra-articular disease manifestation. Patients with RA-ILD experience reduced bacterial diversity and gut bacteriome alterations. However, the gut mycobiome and virome in these patients have been largely neglected. In this study, we performed whole-metagenome shotgun sequencing on fecal samples from 30 patients with RA-ILD, and 30 with RA-non-ILD, and 40 matched healthy controls. The gut bacteriome and mycobiome were explored using a reference-based approach, while the gut virome was profiled based on a nonredundant viral operational taxonomic unit (vOTU) catalog. The results revealed significant alterations in the gut microbiomes of both RA-ILD and RA-non-ILD groups compared with healthy controls. These alterations encompassed changes in the relative abundances of 351 bacterial species, 65 fungal species, and 4,367 vOTUs. Bacteria such as Bifidobacterium longum, Dorea formicigenerans, and Collinsella aerofaciens were enriched in both patient groups. Ruminococcus gnavus (RA-ILD), Gemmiger formicilis, and Ruminococcus bromii (RA-non-ILD) were uniquely enriched. Conversely, Faecalibacterium prausnitzii, Bacteroides spp., and Roseburia inulinivorans showed depletion in both patient groups. Mycobiome analysis revealed depletion of certain fungi, including Saccharomyces cerevisiae and Candida albicans, in patients with RA compared with healthy subjects. Notably, gut virome alterations were characterized by an increase in Siphoviridae and a decrease in Myoviridae, Microviridae, and Autographiviridae in both patient groups. Hence, multikingdom gut microbial signatures showed promise as diagnostic indicators for both RA-ILD and RA-non-ILD. Overall, this study provides comprehensive insights into the fecal virome, bacteriome, and mycobiome landscapes of RA-ILD and RA-non-ILD gut microbiota, thereby offering potential biomarkers for further mechanistic and clinical research.


Subject(s)
Arthritis, Rheumatoid , Bacteria , Feces , Gastrointestinal Microbiome , Lung Diseases, Interstitial , Humans , Lung Diseases, Interstitial/microbiology , Lung Diseases, Interstitial/virology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/microbiology , Feces/microbiology , Feces/virology , Female , Male , Middle Aged , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Aged , Virome , Mycobiome , Adult , Viruses/classification , Viruses/isolation & purification , Viruses/genetics , Fungi/isolation & purification , Fungi/classification
17.
J Med Virol ; 96(7): e29802, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39023095

ABSTRACT

Irritable bowel syndrome (IBS), a chronic functional gastrointestinal disorder, is recognized for its association with alterations in the gut microbiome and metabolome. This study delves into the largely unexplored domain of the gut virome in IBS patients. We conducted a comprehensive analysis of the fecal metagenomic data set from 277 IBS patients and 84 healthy controls to characterize the gut viral community. Our findings revealed a distinct gut virome in IBS patients compared to healthy individuals, marked by significant variances in between-sample diversity and altered abundances of 127 viral operational taxonomic units (vOTUs). Specifically, 111 vOTUs, predominantly belonging to crAss-like, Siphoviridae, Myoviridae, and Quimbyviridae families, were more abundant in IBS patients, whereas the healthy control group exhibited enrichment of 16 vOTUs from multiple families. We also investigated the interplay between the gut virome and bacteriome, identifying a correlation between IBS-enriched bacteria like Klebsiella pneumoniae, Fusobacterium varium, and Ruminococcus gnavus, and the IBS-associated vOTUs. Furthermore, we assessed the potential of gut viral signatures in predicting IBS, achieving a notable area under the receiver operator characteristic curve (AUC) of 0.834. These findings highlight significant shifts in the viral diversity, taxonomic distribution, and functional composition of the gut virome in IBS patients, suggesting the potential role of the gut virome in IBS pathogenesis and opening new avenues for diagnostic and therapeutic strategies targeting the gut virome in IBS management.


Subject(s)
Feces , Gastrointestinal Microbiome , Irritable Bowel Syndrome , Metagenomics , Virome , Humans , Irritable Bowel Syndrome/virology , Irritable Bowel Syndrome/microbiology , Gastrointestinal Microbiome/genetics , Feces/virology , Feces/microbiology , Viruses/classification , Viruses/genetics , Viruses/isolation & purification , Adult , Male , Female , Middle Aged , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Metagenome
18.
Biosens Bioelectron ; 260: 116429, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38838573

ABSTRACT

Developing highly sensitive and specific on-site tests is imperative to strengthen preparedness against future emerging infectious diseases. Here, we describe the construction of a Cas12a-mediated DNAzyme actuator capable of converting the recognition of a specific DNA sequence into an amplified colorimetric signal. To address viral RNA extraction challenges for on-site applications, we developed a rapid and efficient method capable of lysing the viral particles, preserving the released viral RNA, and concentrating the viral RNA. Integration of the DNAzyme actuator with the viral RNA extraction method and loop-mediated isothermal amplification enables a streamlined colorimetric assay for highly sensitive colorimetric detection of respiratory RNA viruses in gargle and saliva. This assay can detect as few as 83 viral particles/100 µL in gargle and 166 viral particles/100 µL in saliva. The entire assay, from sample processing to visual detection, was completed within 1 h at a single controlled temperature. We validated the assay by detecting SARS-CoV-2 in 207 gargle and saliva samples, achieving a clinical sensitivity of 96.3 % and specificity of 100%. The assay is adaptable for detecting specific nucleic acid sequences in other pathogens and is suitable for resource-limited settings.


Subject(s)
Biosensing Techniques , Colorimetry , DNA, Catalytic , Nucleic Acid Amplification Techniques , RNA, Viral , SARS-CoV-2 , Saliva , Colorimetry/methods , RNA, Viral/isolation & purification , RNA, Viral/genetics , SARS-CoV-2/isolation & purification , SARS-CoV-2/genetics , DNA, Catalytic/chemistry , Biosensing Techniques/methods , Saliva/virology , Saliva/chemistry , Humans , Nucleic Acid Amplification Techniques/methods , COVID-19/virology , COVID-19/diagnosis , CRISPR-Associated Proteins/isolation & purification , CRISPR-Associated Proteins/chemistry , Endodeoxyribonucleases/chemistry , Limit of Detection , Feces/virology , Feces/chemistry , Bacterial Proteins , Molecular Diagnostic Techniques
19.
BMC Genomics ; 25(1): 629, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38914944

ABSTRACT

BACKGROUND: Virome studies on birds, including chickens are relatively scarce, particularly from the African continent. Despite the continuous evolution of RNA viruses and severe losses recorded in poultry from seasonal viral outbreaks, the information on RNA virome composition is even scantier as a result of their highly unstable nature, genetic diversity, and difficulties associated with characterization. Also, information on factors that may modulate the occurrence of some viruses in birds is limited, particularly for domesticated birds. Viral metagenomics through advancements in sequencing technologies, has enabled the characterization of the entire virome of diverse host species using various samples. METHODS: The complex RNA viral constituents present in 27 faecal samples of asymptomatic chickens from a South African farm collected at 3-time points from two independent seasons were determined, and the impact of the chicken's age and collection season on viral abundance and diversity was further investigated. The study utilized the non-invasive faecal sampling method, mRNA viral targeted enrichment steps, a whole transcriptome amplification strategy, Illumina sequencing, and bioinformatics tools. RESULTS: The results obtained revealed a total of 48 viral species spanning across 11 orders, 15 families and 21 genera. Viral RNA families such as Coronaviridae, Picornaviridae, Reoviridae, Astroviridae, Caliciviridae, Picorbirnaviridae and Retroviridae were abundant, among which picornaviruses, demonstrated a 100% prevalence across the three age groups (2, 4 and 7 weeks) and two seasons (summer and winter) of the 27 faecal samples investigated. A further probe into the extent of variation between the different chicken groups investigated indicated that viral diversity and abundance were significantly influenced by age (P = 0.01099) and season (P = 0.00099) between chicken groups, while there was no effect on viral shedding within samples in a group (alpha diversity) for age (P = 0.146) and season (P = 0.242). CONCLUSION: The presence of an exceedingly varied chicken RNA virome, encompassing avian, mammalian, fungal, and dietary-associated viruses, underscores the complexities inherent in comprehending the causation, dynamics, and interspecies transmission of RNA viruses within the investigated chicken population. Hence, chickens, even in the absence of discernible symptoms, can harbour viruses that may exhibit opportunistic, commensal, or pathogenic characteristics.


Subject(s)
Chickens , Feces , Metagenomics , RNA, Viral , Virome , Animals , Chickens/virology , South Africa/epidemiology , Feces/virology , Virome/genetics , Metagenomics/methods , RNA, Viral/genetics , RNA Viruses/genetics , RNA Viruses/classification , RNA Viruses/isolation & purification , Farms , Metagenome , Seasons
20.
Nat Commun ; 15(1): 4704, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830845

ABSTRACT

Metabolic syndrome encompasses amongst other conditions like obesity and type-2 diabetes and is associated with gut microbiome (GM) dysbiosis. Fecal microbiota transplantation (FMT) has been explored to treat metabolic syndrome by restoring the GM; however, concerns on accidentally transferring pathogenic microbes remain. As a safer alternative, fecal virome transplantation (FVT, sterile-filtrated feces) has the advantage over FMT in that mainly bacteriophages are transferred. FVT from lean male donors have shown promise in alleviating the metabolic effects of high-fat diet in a preclinical mouse study. However, FVT still carries the risk of eukaryotic viral infections. To address this, recently developed methods are applied for removing or inactivating eukaryotic viruses in the viral component of FVT. Modified FVTs are compared with unmodified FVT and saline in a diet-induced obesity model on male C57BL/6 N mice. Contrasted with obese control, mice administered a modified FVT (nearly depleted for eukaryotic viruses) exhibits enhanced blood glucose clearance but not weight loss. The unmodified FVT improves liver pathology and reduces the proportions of immune cells in the adipose tissue with a non-uniform response. GM analysis suggests that bacteriophage-mediated GM modulation influences outcomes. Optimizing these approaches could lead to the development of safe bacteriophage-based therapies targeting metabolic syndrome through GM restoration.


Subject(s)
Diet, High-Fat , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Metabolic Syndrome , Mice, Inbred C57BL , Mice, Obese , Obesity , Virome , Animals , Male , Metabolic Syndrome/therapy , Obesity/therapy , Mice , Diet, High-Fat/adverse effects , Dysbiosis/therapy , Feces/virology , Feces/microbiology , Bacteriophages/physiology , Blood Glucose/metabolism , Disease Models, Animal , Liver/pathology , Liver/metabolism , Adipose Tissue
SELECTION OF CITATIONS
SEARCH DETAIL
...