Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
J Appl Microbiol ; 134(1)2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36626787

ABSTRACT

Omics research inevitably involves the collection and analysis of big data, which can only be handled by automated approaches. Here we point out that the analysis of big data in the field of genomics dictates certain requirements, such as specialized software, quality control of input data, and simplification for visualization of the results. The latter results in a loss of information, as is exemplified for phylogenetic trees. Clear communication of big data analyses can be enhanced by novel visualization strategies. The interpretation of findings is sometimes hampered when dedicated analytical tools are not fully understood by microbiologists, while the researchers performing these analyses may not have a full overview of the biology of the microbes under study. These issues are illustrated here, using SARS-Cov-2 and Salmonella enterica as zoonotic examples. Whereas in scientific communications jargon should be avoided or explained, nomenclature to group similar organisms and distinguish these from more distant relatives is not only essential, but also influences the interpretation of results. Unfortunately, changes in taxonomically accepted names are now so frequent that they hamper rather than assist research, as is illustrated with difficulties of microbiome studies. Nomenclature to group viral isolates, as is done for SARS-Cov2, is also not without difficulties. Some weaknesses in current omics research stem from poor quality of data or biased databases, and problems can be magnified by machine learning approaches. Moreover, the overall opus of scientific publications can now be considered "big data", as is illustrated by the avalanche of COVID-19-related publications. The peer-review model of scientific publishing is only barely coping with this novel situation, resulting in retractions and the publication of bogus works. The avalanche of scientific publications that originated from the current pandemic can obstruct literature searches, and this will unfortunately continue over time.


Subject(s)
COVID-19 , Animals , Humans , SARS-CoV-2/genetics , Phylogeny , RNA, Viral , Genomics , Zoonoses
2.
J Appl Microbiol ; 133(6): 3690-3698, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36074056

ABSTRACT

AIMS: The current Monkeypox virus (MPX) outbreak is not only the largest known outbreak to date caused by a strain belonging to the West-African clade, but also results in remarkably different clinical and epidemiological features compared to previous outbreaks of this virus. Here, we consider the possibility that mutations in the viral genome may be responsible for its changed characteristics. METHODS AND RESULTS: Six genome sequences of isolates from the current outbreak were compared to five genomes of isolates from the 2017 outbreak in Nigeria and to two historic genomes, all belonging to the West-African clade. We report differences that are consistently present in the 2022 isolates but not in the others. Although some variation in repeat units was observed, only two were consistently found in the 2022 genomes only, and these were located in intergenic regions. A total of 55 single nucleotide polymorphisms were consistently present in the 2022 isolates compared to the 2017 isolates. Of these, 25 caused an amino acid substitution in a predicted protein. CONCLUSIONS: The nature of the substitution and the annotation of the affected protein identified potential candidates that might affect the virulence of the virus. These included the viral DNA helicase and transcription factors. SIGNIFICANCE: This bioinformatic analysis provides guidance for wet-lab research to identify changed properties of the MPX.


Subject(s)
Disease Outbreaks , Monkeypox virus , Monkeypox virus/genetics , Nigeria/epidemiology , Genome, Viral/genetics , DNA, Viral
3.
FEMS Microbiol Rev ; 46(3)2022 05 06.
Article in English | MEDLINE | ID: mdl-35076068

ABSTRACT

The genomic diversity of SARS-CoV-2 is the result of a relatively low level of spontaneous mutations introduced during viral replication. With millions of SARS-CoV-2 genome sequences now available, we can begin to assess the overall genetic repertoire of this virus. We find that during 2020, there was a global wave of one variant that went largely unnoticed, possibly because its members were divided over several sublineages (B.1.177 and sublineages B.1.177.XX). We collectively call this Janus, and it was eventually replaced by the Alpha (B.1.1.7) variant of concern (VoC), next replaced by Delta (B.1.617.2), which itself might soon be replaced by a fourth pandemic wave consisting of Omicron (B.1.1.529). We observe that splitting up and redefining variant lineages over time, as was the case with Janus and is now happening with Alpha, Delta and Omicron, is not helpful to describe the epidemic waves spreading globally. Only ∼5% of the 30 000 nucleotides of the SARS-CoV-2 genome are found to be variable. We conclude that a fourth wave of the pandemic with the Omicron variant might not be that different from other VoCs, and that we may already have the tools in hand to effectively deal with this new VoC.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/epidemiology , Humans , Mutation , Pandemics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics
4.
Int J Mol Sci ; 22(18)2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34575864

ABSTRACT

The importance of a healthy microbiome cannot be overemphasized. Disturbances in its composition can lead to a variety of symptoms that can extend to other organs. Likewise, acute or chronic conditions in other organs can affect the composition and physiology of the gut microbiome. Here, we discuss interorgan communication along the gut-lung axis, as well as interactions between lung and coronary heart diseases and between cardiovascular disease and the gut microbiome. This triangle of organs, which also affects the clinical outcome of COVID-19 infections, is connected by means of numerous receptors and effectors, including immune cells and immune-modulating factors such as short chain fatty acids (SCFA) and trimethlamine-N-oxide (TMAO). The gut microbiome plays an important role in each of these, thus affecting the health of the lungs and the heart, and this interplay occurs in both directions. The gut microbiome can be influenced by the oral uptake of probiotics. With an improved understanding of the mechanisms responsible for interorgan communication, we can start to define what requirements an 'ideal' probiotic should have and its role in this triangle.


Subject(s)
COVID-19 , Coronary Disease , Gastrointestinal Microbiome/drug effects , Lung Diseases , Probiotics/administration & dosage , Animals , COVID-19/microbiology , COVID-19/pathology , Coronary Disease/microbiology , Coronary Disease/pathology , Humans , Lung Diseases/microbiology , Lung Diseases/pathology
5.
Commun Biol ; 4(1): 117, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33500552

ABSTRACT

In this study, more than one hundred thousand Escherichia coli and Shigella genomes were examined and classified. This is, to our knowledge, the largest E. coli genome dataset analyzed to date. A Mash-based analysis of a cleaned set of 10,667 E. coli genomes from GenBank revealed 14 distinct phylogroups. A representative genome or medoid identified for each phylogroup was used as a proxy to classify 95,525 unassembled genomes from the Sequence Read Archive (SRA). We find that most of the sequenced E. coli genomes belong to four phylogroups (A, C, B1 and E2(O157)). Authenticity of the 14 phylogroups is supported by several different lines of evidence: phylogroup-specific core genes, a phylogenetic tree constructed with 2613 single copy core genes, and differences in the rates of gene gain/loss/duplication. The methodology used in this work is able to reproduce known phylogroups, as well as to identify previously uncharacterized phylogroups in E. coli species.


Subject(s)
Escherichia coli/classification , Escherichia coli/genetics , Genome, Bacterial , Computational Biology/methods , Escherichia coli Proteins/genetics , Genetic Speciation , Genomics/methods , Phylogeny , Sequence Analysis, DNA , Shigella/classification , Shigella/genetics
6.
Nucleic Acids Res ; 49(2): e7, 2021 01 25.
Article in English | MEDLINE | ID: mdl-32710622

ABSTRACT

Traditional epitranscriptomics relies on capturing a single RNA modification by antibody or chemical treatment, combined with short-read sequencing to identify its transcriptomic location. This approach is labor-intensive and may introduce experimental artifacts. Direct sequencing of native RNA using Oxford Nanopore Technologies (ONT) can allow for directly detecting the RNA base modifications, although these modifications might appear as sequencing errors. The percent Error of Specific Bases (%ESB) was higher for native RNA than unmodified RNA, which enabled the detection of ribonucleotide modification sites. Based on the %ESB differences, we developed a bioinformatic tool, epitranscriptional landscape inferring from glitches of ONT signals (ELIGOS), that is based on various types of synthetic modified RNA and applied to rRNA and mRNA. ELIGOS is able to accurately predict known classes of RNA methylation sites (AUC > 0.93) in rRNAs from Escherichiacoli, yeast, and human cells, using either unmodified in vitro transcription RNA or a background error model, which mimics the systematic error of direct RNA sequencing as the reference. The well-known DRACH/RRACH motif was localized and identified, consistent with previous studies, using differential analysis of ELIGOS to study the impact of RNA m6A methyltransferase by comparing wild type and knockouts in yeast and mouse cells. Lastly, the DRACH motif could also be identified in the mRNA of three human cell lines. The mRNA modification identified by ELIGOS is at the level of individual base resolution. In summary, we have developed a bioinformatic software package to uncover native RNA modifications.


Subject(s)
Computational Biology/methods , High-Throughput Nucleotide Sequencing , RNA Processing, Post-Transcriptional , RNA-Seq , Scientific Experimental Error , Software , Adenine/analogs & derivatives , Adenine/analysis , Animals , Cell Line , Escherichia coli/genetics , Humans , Meiosis , Methyltransferases/deficiency , Methyltransferases/metabolism , Mice , Mice, Knockout , Nucleotide Motifs , RNA, Bacterial/genetics , RNA, Fungal/genetics , RNA, Messenger/genetics , RNA, Ribosomal/genetics , ROC Curve , Saccharomyces cerevisiae/genetics , Sequence Analysis, DNA , Templates, Genetic , Transcription, Genetic
7.
Eur J Microbiol Immunol (Bp) ; 10(1): 11-19, 2020 Apr 07.
Article in English | MEDLINE | ID: mdl-32363034

ABSTRACT

Probiotic Escherichia coli strain Nissle 1917 (EcN) has a long history of safe use. However, the recently discovered presence of a pks locus in its genome presumably producing colibactin has questioned its safety, as colibactin has been implicated in genotoxicity. Here, we assess the genotoxic potential of EcN. Metabolic products were tested in vitro by the Ames test, a mutagenicity assay developed to detect point mutation-inducing activity. Live EcN were tested by an adapted Ames test. Neither the standard nor the adapted Ames test resulted in increased numbers of revertant colonies, indicating that EcN metabolites or viable cells lacked mutagenic activity. The in vivo Mammalian Alkaline Comet Assay (the gold standard for detecting DNA-strand breaks) was used to determine potentially induced DNA-strand breaks in cells of the gastro-intestinal tract of rats orally administered with viable EcN. Bacteria were given at 109-1011 colony forming units (CFU) per animal by oral gavage on 2 consecutive days and daily for a period of 28 days to 5 rats per group. No significant differences compared to negative controls were found. These results demonstrate that EcN does not induce DNA-strand breaks and does not have any detectable genotoxic potential in the test animals.

8.
Microorganisms ; 8(2)2020 Feb 12.
Article in English | MEDLINE | ID: mdl-32059450

ABSTRACT

Recently, Zimmer and Dorea published a communication on the enumeration of Escherichia coli in probiotic products containing this species [...].

9.
Microbiologyopen ; 9(2): e973, 2020 02.
Article in English | MEDLINE | ID: mdl-31742930

ABSTRACT

The intraspecies genomic diversity of the single-strand RNA (+) virus species hepatitis A virus (Hepatovirus), hepatitis C virus (Hepacivirus), and hepatitis E virus (Orthohepevirus) was compared. These viral species all can cause liver inflammation (hepatitis), but share no gene similarity. The codon usage of human hepatitis A virus (HAV) is suboptimal for replication in its host, a characteristic it shares with taxonomically related rodent, simian, and bat hepatitis A virus species. We found this codon usage to be strikingly similar to that of Triatoma virus that infects blood-sucking kissing bugs. The codon usage of that virus is well adapted to its insect host. The codon usage of HAV is also similar to other invertebrate viruses of various taxonomic families. An evolutionary ancestor of HAV and related virus species is hypothesized to be an insect virus that underwent a host jump to infect mammals. The similarity between HAV and invertebrate viruses goes beyond codon usage, as they also share amino acid composition characteristics, while not sharing direct sequence homology. In contrast, hepatitis C virus and hepatitis E virus are highly similar in codon usage preference, nucleotide composition, and amino acid composition, and share these characteristics with Human pegivirus A, West Nile virus, and Zika virus. We present evidence that these observations are only partly explained by differences in nucleotide composition of the complete viral codon regions. We consider the combination of nucleotide composition, amino acid composition, and codon usage preference suitable to provide information on possible evolutionary similarities between distant virus species that cannot be investigated by phylogeny.


Subject(s)
Evolution, Molecular , Genome, Viral , Genomics , Hepacivirus/genetics , Hepatitis A virus/genetics , Hepatitis E virus/genetics , Codon , Genomics/methods , Hepacivirus/classification , Hepatitis A virus/classification , Hepatitis E virus/classification , Humans , Phylogeny
10.
Front Microbiol ; 10: 857, 2019.
Article in English | MEDLINE | ID: mdl-31080442

ABSTRACT

To achieve maximum transmission chain tracking in the current Ebola outbreak, whole genome sequencing (WGS) has been proposed to provide optimal information. However, WGS remains a costly and time-intensive procedure that is poorly suited for the large numbers of samples being generated, especially under severe time and work-environment constraints as in the present DRC outbreak. To better prepare for future outbreaks, where an apparent single outbreak may actually represent overlapping outbreaks caused by independent variants, and where rapid identification of emerging new transmission chains will be essential, a more practical method would be to amplify and sequence genomic areas that reveal the highest information to differentiate EBOV variants. We have identified four highly informative polymorphism PCR sequencing targets, suitable for rapid tracing of transmission chains and identification of new sources of Ebola outbreaks, an approach which will be far more practical in the field than WGS.

11.
Eur Heart J ; 40(14): 1107-1112, 2019 04 07.
Article in English | MEDLINE | ID: mdl-30753448

ABSTRACT

Cardiovascular disease (CVD) rates in adulthood are high in premature infants; unfortunately, the underlying mechanisms are not well defined. In this review, we discuss potential pathways that could lead to CVD in premature babies. Studies show intense oxidant stress and inflammation at tissue levels in these neonates. Alterations in lipid profile, foetal epigenomics, and gut microbiota in these infants may also underlie the development of CVD. Recently, probiotic bacteria, such as the mucin-degrading bacterium Akkermansia muciniphila have been shown to reduce inflammation and prevent heart disease in animal models. All this information might enable scientists and clinicians to target pathways to act early to curtail the adverse effects of prematurity on the cardiovascular system. This could lead to primary and secondary prevention of CVD and improve survival among preterm neonates later in adult life.


Subject(s)
Cardiovascular Diseases/physiopathology , Premature Birth/physiopathology , Atherosclerosis/physiopathology , Cytokines/metabolism , Dyslipidemias/physiopathology , Endothelium, Vascular/physiopathology , Epigenesis, Genetic/physiology , Gastrointestinal Microbiome/physiology , Humans , Inflammation/metabolism , Inflammation/physiopathology , Metabolic Syndrome/physiopathology , Nitric Oxide/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/physiology
12.
Eur J Microbiol Immunol (Bp) ; 8(3): 63-69, 2018 Sep 28.
Article in English | MEDLINE | ID: mdl-30345085

ABSTRACT

The fever-inducing effect of lipopolysaccharides (LPS) is well known, and human blood is extremely responsive to this pyrogen. Recently, the safety of LPS-containing food supplements and probiotic drugs as immune-stimulants has been questioned, although these products are orally taken and do not reach the bloodstream undigested. The concerns are understandable, as endotoxaemia is a pathological condition, but the oral uptake of probiotic products containing LPS or Gram-negative bacteria does not pose a health risk, based on the available scientific evidence, as is reviewed here. The available methods developed to detect LPS and other pyrogens are mostly used for quality control of parentally applied therapeuticals. Their outcome varies considerably when applied to food supplements, as demonstrated in a simple comparative experiment. Products containing different Escherichia coli strains can result in vastly different results on their LPS content, depending on the method of testing. This is an inherent complication to pyrogen testing, which hampers the communication that the LPS content of food supplements is not a safety concern.

13.
Emerg Infect Dis ; 24(9)2018 09.
Article in English | MEDLINE | ID: mdl-29985788

ABSTRACT

We sequenced the virus genomes from 3 pregnant women in Thailand with Zika virus diagnoses. All had infections with the Asian lineage. The woman infected at gestational week 9, and not those infected at weeks 20 and 24, had a fetus with microcephaly. Asian lineage Zika viruses can cause microcephaly.


Subject(s)
Microcephaly/diagnosis , Pregnancy Complications, Infectious , Zika Virus Infection , Zika Virus/isolation & purification , Female , Humans , Infant, Newborn , Microcephaly/etiology , Pregnancy , Pregnancy Trimester, First , Thailand , Zika Virus/genetics
14.
Crit Rev Microbiol ; 44(5): 619-632, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29909724

ABSTRACT

To the multiple factors that may eventually result in colorectal cancer (CRC), strains of E. coli have now been added, in particular strains producing colibactin from their polyketide synthesis (pks) locus. The evidence and mechanistic explanations for this unfortunate effect of what is in most cases a harmless commensal are discussed in the first part of this review. In the second part, observations are presented and discussed that do not fit with the hypothesis that colibactin-producing E. coli produce CRC. The last part of this review is reserved for an alternative explanation of the function of this enigmatic colibactin, a toxin that has not yet been isolated. It is hypothesized that E. coli preferentially colonizes cancerous lesions as an effect rather than a cause and that colibactin production provides a selective advantage to compete with other bacteria.


Subject(s)
Colorectal Neoplasms/microbiology , Escherichia coli Infections/microbiology , Escherichia coli/metabolism , Animals , Escherichia coli/genetics , Escherichia coli/growth & development , Humans , Peptides/metabolism , Polyketides/metabolism
15.
Microb Ecol ; 76(3): 801-813, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29445826

ABSTRACT

Infections due to Clostridioides difficile (previously known as Clostridium difficile) are a major problem in hospitals, where cases can be caused by community-acquired strains as well as by nosocomial spread. Whole genome sequences from clinical samples contain a lot of information but that needs to be analyzed and compared in such a way that the outcome is useful for clinicians or epidemiologists. Here, we compare 663 public available complete genome sequences of C. difficile using average amino acid identity (AAI) scores. This analysis revealed that most of these genomes (640, 96.5%) clearly belong to the same species, while the remaining 23 genomes produce four distinct clusters within the Clostridioides genus. The main C. difficile cluster can be further divided into sub-clusters, depending on the chosen cutoff. We demonstrate that MLST, either based on partial or full gene-length, results in biased estimates of genetic differences and does not capture the true degree of similarity or differences of complete genomes. Presence of genes coding for C. difficile toxins A and B (ToxA/B), as well as the binary C. difficile toxin (CDT), was deduced from their unique PfamA domain architectures. Out of the 663 C. difficile genomes, 535 (80.7%) contained at least one copy of ToxA or ToxB, while these genes were missing from 128 genomes. Although some clusters were enriched for toxin presence, these genes are variably present in a given genetic background. The CDT genes were found in 191 genomes, which were restricted to a few clusters only, and only one cluster lacked the toxin A/B genes consistently. A total of 310 genomes contained ToxA/B without CDT (47%). Further, published metagenomic data from stools were used to assess the presence of C. difficile sequences in blinded cases of C. difficile infection (CDI) and controls, to test if metagenomic analysis is sensitive enough to detect the pathogen, and to establish strain relationships between cases from the same hospital. We conclude that metagenomics can contribute to the identification of CDI and can assist in characterization of the most probable causative strain in CDI patients.


Subject(s)
Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Genome, Bacterial , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Toxins/metabolism , Clostridioides difficile/chemistry , Clostridioides difficile/classification , Clostridium Infections/microbiology , Gene Dosage , Humans , Molecular Sequence Data , Multilocus Sequence Typing , Phylogeny , Sequence Homology, Amino Acid
16.
Nat Commun ; 8(1): 698, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28951596

ABSTRACT

Colonization of body epithelial surfaces with a highly specific microbial community is a fundamental feature of all animals, yet the underlying mechanisms by which these communities are selected and maintained are not well understood. Here, we show that sensory and ganglion neurons in the ectodermal epithelium of the model organism hydra (a member of the animal phylum Cnidaria) secrete neuropeptides with antibacterial activity that may shape the microbiome on the body surface. In particular, a specific neuropeptide, which we call NDA-1, contributes to the reduction of Gram-positive bacteria during early development and thus to a spatial distribution of the main colonizer, the Gram-negative Curvibacter sp., along the body axis. Our findings warrant further research to test whether neuropeptides secreted by nerve cells contribute to the spatial structure of microbial communities in other organisms.Certain neuropeptides, in addition to their neuromodulatory functions, display antibacterial activities of unclear significance. Here, the authors show that a secreted neuropeptide modulates the distribution of bacterial communities on the body surface during development of the model organism Hydra.


Subject(s)
Anti-Bacterial Agents/metabolism , Hydra/microbiology , Microbiota , Neurons/metabolism , Neuropeptides/metabolism , Animals , Comamonadaceae , Ectoderm/cytology , Ectoderm/metabolism , Epithelium/metabolism , Gram-Positive Bacteria , Hydra/growth & development , Hydra/metabolism
17.
Genome Announc ; 5(32)2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28798162

ABSTRACT

We present here the complete genome sequences of three mumps virus (MuV) strains isolated from patients who tested positive for the mumps virus during a mumps outbreak in Springdale, AR (USA), in 2016. The virus genomes, sequenced with Oxford Nanopore technology, belong to genotype G and have an average length of 15,342 nucleotides (nt).

18.
BMC Bioinformatics ; 18(Suppl 14): 471, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29297281

ABSTRACT

BACKGROUND: Zika virus (ZIKV) is an emerging human pathogen. Since its arrival in the Western hemisphere, from Africa via Asia, it has become a serious threat to pregnant women, causing microcephaly and other neuropathies in developing fetuses. The mechanisms behind these teratogenic effects are unknown, although epidemiological evidence suggests that microcephaly is not associated with the original, African lineage of ZIKV. The sequences of 196 published ZIKV genomes were used to assess whether recently proposed mechanistic explanations for microcephaly are supported by molecular level changes that may have increased its virulence since the virus left Africa. For this we performed phylogenetic, recombination, adaptive evolution and tetramer frequency analyses, and compared protein sequences for the presence of protease cleavage sites, Pfam domains, glycosylation sites, signal peptides, trans-membrane protein domains, and phosphorylation sites. RESULTS: Recombination events within or between Asian and Brazilian lineages were not observed, and likewise there were no differences in protease cleavage, glycosylation sites, signal peptides or trans-membrane domains between African and Brazilian strains. The frequency of Retinoic Acid Response Element (RARE) sequences was increased in Brazilian strains. Genetic adaptation was also apparent by tetramer signatures that had undergone major changes in the past but has stabilized in the Brazilian lineage despite subsequent geographic spread, suggesting the viral population presently propagates in the same host species in various regions. Evidence for selection pressure was recognized for several amino acid sites in the Brazilian lineage compared to the African lineage, mainly in nonstructural proteins, especially protein NS4B. A number of these positively selected mutations resulted in an increased potential to be phosphorylated in the Brazilian lineage compared to the African linage, which may have increased their potential to interfere with neural fetal development. CONCLUSIONS: ZIKV seems to have adapted to a limited number of hosts, including humans, during which its virulence increased. Its protein NS4B, together with NS4A, has recently been shown to inhibit Akt-mTOR signaling in human fetal neural stem cells, a key pathway for brain development. We hypothesize that positive selection of novel phosphorylation sites in the protein NS4B of the Brazilian lineage could interfere with phosphorylation of Akt and mTOR, impairing Akt-mTOR signaling and this may result in an increased risk for developmental neuropathies.


Subject(s)
Genome, Viral , Microcephaly/virology , Zika Virus/genetics , Zika Virus/physiology , Adaptation, Physiological/genetics , Africa , Asia , Base Sequence , Brazil , Cell Line , Codon/genetics , Female , Genetic Variation , Host-Pathogen Interactions/genetics , Humans , Microcephaly/immunology , Phosphorylation , Phylogeny , Pregnancy , RNA Stability/genetics , Recombination, Genetic/genetics , Selection, Genetic , Virulence/genetics , Zika Virus/pathogenicity , Zika Virus Infection/immunology , Zika Virus Infection/virology
19.
Front Microbiol ; 7: 1528, 2016.
Article in English | MEDLINE | ID: mdl-27729906

ABSTRACT

Resistance of Staphylococcus species to quaternary ammonium compounds, frequently used as disinfectants and biocides, can be attributed to qac genes. Most qac gene products belong to the Small Multidrug Resistant (SMR) protein family, and are often encoded by rolling-circle (RC) replicating plasmids. Four classes of SMR-type qac gene families have been described in Staphylococcus species: qacC, qacG, qacJ, and qacH. Within their class, these genes are highly conserved, but qacC genes are extremely conserved, although they are found in variable plasmid backgrounds. The lower degree of sequence identity of these plasmids compared to the strict nucleotide conservation of their qacC means that this gene has recently spread. In the absence of insertion sequences or other genetic elements explaining the mobility, we sought for an explanation of mobilization by sequence comparison. Publically available sequences of qac genes, their flanking genes and the replication gene that is invariably present in RC-plasmids were compared to reconstruct the evolutionary history of these plasmids and to explain the recent spread of qacC. Here we propose a new model that explains how qacC is mobilized and transferred to acceptor RC-plasmids without assistance of other genes, by means of its location in between the Double Strand replication Origin (DSO) and the Single-Strand replication Origin (SSO). The proposed mobilization model of this DSO-qacC-SSO element represents a novel mechanism of gene mobilization in RC-plasmids, which has also been employed by other genes, such as lnuA (conferring lincomycin resistance). The proposed gene mobility has aided to the wide spread of clinically relevant resistance genes in Staphylococcus populations.

20.
Eur J Microbiol Immunol (Bp) ; 6(3): 147-161, 2016 Sep 29.
Article in English | MEDLINE | ID: mdl-27766164

ABSTRACT

A century ago, Alfred Nissle discovered that intentional intake of particular strains of Escherichia coli could treat patients suffering from infectious diseases. Since then, one of these strains became the most frequently used probiotic E. coli in research and was applied to a variety of human conditions. Here, properties of that E. coli Nissle 1917 strain are compared with other commercially available E. coli probiotic strains, with emphasis on their human applications. A literature search formed the basis of a summary of research findings reported for the probiotics Mutaflor, Symbioflor 2, and Colinfant. The closest relatives of the strains in these products are presented, and their genetic content, including the presence of virulence, genes is discussed. A similarity to pathogenic strains causing urinary tract infections is noticeable. Historic trends in research of probiotics treatment for particular human conditions are identified. The future of probiotic E. coli may lay in what Alfred Nissle originally discovered: to treat gastrointestinal infections, which nowadays are often caused by antibiotic-resistant pathogens.

SELECTION OF CITATIONS
SEARCH DETAIL
...