Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Euro Surveill ; 29(27)2024 Jul.
Article in English | MEDLINE | ID: mdl-38967012

ABSTRACT

During the summer of 2023, the European Region experienced a limited resurgence of mpox cases following the substantial outbreak in 2022. This increase was characterised by asynchronous and bimodal increases, with countries experiencing peaks at different times. The demographic profile of cases during the resurgence was largely consistent with those reported previously. All available sequences from the European Region belonged to clade IIb. Sustained efforts are crucial to control and eventually eliminate mpox in the European Region.


Subject(s)
Disease Outbreaks , Phylogeny , Humans , Europe/epidemiology , Male , Female , Adult , Middle Aged , Adolescent , Young Adult , Child , Aged , Population Surveillance , Child, Preschool , Incidence
3.
EFSA J ; 21(2): e07822, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36860662

ABSTRACT

The epidemiological situation of SARS-CoV-2 in humans and animals is continually evolving. To date, animal species known to transmit SARS-CoV-2 are American mink, raccoon dog, cat, ferret, hamster, house mouse, Egyptian fruit bat, deer mouse and white-tailed deer. Among farmed animals, American mink have the highest likelihood to become infected from humans or animals and further transmit SARS-CoV-2. In the EU, 44 outbreaks were reported in 2021 in mink farms in seven MSs, while only six in 2022 in two MSs, thus representing a decreasing trend. The introduction of SARS-CoV-2 into mink farms is usually via infected humans; this can be controlled by systematically testing people entering farms and adequate biosecurity. The current most appropriate monitoring approach for mink is the outbreak confirmation based on suspicion, testing dead or clinically sick animals in case of increased mortality or positive farm personnel and the genomic surveillance of virus variants. The genomic analysis of SARS-CoV-2 showed mink-specific clusters with a potential to spill back into the human population. Among companion animals, cats, ferrets and hamsters are those at highest risk of SARS-CoV-2 infection, which most likely originates from an infected human, and which has no or very low impact on virus circulation in the human population. Among wild animals (including zoo animals), mostly carnivores, great apes and white-tailed deer have been reported to be naturally infected by SARS-CoV-2. In the EU, no cases of infected wildlife have been reported so far. Proper disposal of human waste is advised to reduce the risks of spill-over of SARS-CoV-2 to wildlife. Furthermore, contact with wildlife, especially if sick or dead, should be minimised. No specific monitoring for wildlife is recommended apart from testing hunter-harvested animals with clinical signs or found-dead. Bats should be monitored as a natural host of many coronaviruses.

5.
Proc Natl Acad Sci U S A ; 119(23): e2122386119, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35648835

ABSTRACT

Pneumococcal conjugate vaccines (PCVs) used in childhood vaccination programs have resulted in replacement of vaccine-type with nonvaccine-type pneumococci in carriage and invasive pneumococcal disease (IPD). A vaccine based on highly conserved and protective pneumococcal antigens is urgently needed. Here, we performed intranasal immunization of mice with pneumococcal membrane particles (MPs) to mimic natural nasopharyngeal immunization. MP immunization gave excellent serotype-independent protection against IPD that was antibody dependent but independent of the cytotoxin pneumolysin. Using Western blotting, immunoprecipitation, mass spectrometry, and different bacterial mutants, we identified the conserved lipoproteins MalX and PrsA as the main antigens responsible for cross-protection. Additionally, we found that omitting the variable surface protein and vaccine candidate PspA from MPs enhanced protective immune responses to the conserved proteins. Our findings suggest that MPs containing MalX and PrsA could serve as a platform for pneumococcal vaccine development targeting the elderly and immunocompromised.


Subject(s)
Bacterial Proteins , Lipoproteins , Membrane Proteins , Membrane Transport Proteins , Pneumococcal Infections , Pneumococcal Vaccines , Administration, Intranasal , Animals , Bacterial Proteins/immunology , Cell Membrane/immunology , Conserved Sequence , Cross Reactions , Humans , Immunization/methods , Lipoproteins/immunology , Membrane Proteins/immunology , Membrane Transport Proteins/immunology , Mice , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Pneumococcal Vaccines/immunology , Serogroup , Streptococcus pneumoniae/immunology
6.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34785593

ABSTRACT

Emerging antibiotic resistance demands identification of novel antibacterial compound classes. A bacterial whole-cell screen based on pneumococcal autolysin-mediated lysis induction was developed to identify potential bacterial cell wall synthesis inhibitors. A hit class comprising a 1-amino substituted tetrahydrocarbazole (THCz) scaffold, containing two essential amine groups, displayed bactericidal activity against a broad range of gram-positive and selected gram-negative pathogens in the low micromolar range. Mode of action studies revealed that THCz inhibit cell envelope synthesis by targeting undecaprenyl pyrophosphate-containing lipid intermediates and thus simultaneously inhibit peptidoglycan, teichoic acid, and polysaccharide capsule biosynthesis. Resistance did not readily develop in vitro, and the ease of synthesizing and modifying these small molecules, as compared to natural lipid II-binding antibiotics, makes THCz promising scaffolds for development of cell wall-targeting antimicrobials.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Cell Wall/chemistry , Cell Wall/drug effects , Lipids/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Drug Resistance, Bacterial/drug effects , Microbial Sensitivity Tests , N-Acetylmuramoyl-L-alanine Amidase , Peptidoglycan/biosynthesis , Polyisoprenyl Phosphates , Streptococcus pneumoniae/drug effects , Teichoic Acids/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives
7.
mBio ; 11(6)2020 12 15.
Article in English | MEDLINE | ID: mdl-33323510

ABSTRACT

Streptococcus pneumoniae, a major cause of pneumonia, sepsis, and meningitis worldwide, has the nasopharynges of small children as its main ecological niche. Depletion of pneumococci from this niche would reduce the disease burden and could be achieved using small molecules with narrow-spectrum antibacterial activity. We identified the alkylated dicyclohexyl carboxylic acid 2CCA-1 as a potent inducer of autolysin-mediated lysis of S. pneumoniae, while having low activity against Staphylococcus aureus 2CCA-1-resistant strains were found to have inactivating mutations in fakB3, known to be required for uptake of host polyunsaturated fatty acids, as well as through inactivation of the transcriptional regulator gene fabT, vital for endogenous, de novo fatty acid synthesis regulation. Structure activity relationship exploration revealed that, besides the central dicyclohexyl group, the fatty acid-like structural features of 2CCA-1 were essential for its activity. The lysis-inducing activity of 2CCA-1 was considerably more potent than that of free fatty acids and required growing bacteria, suggesting that 2CCA-1 needs to be metabolized to exert its antimicrobial activity. Total lipid analysis of 2CCA-1 treated bacteria identified unique masses that were modeled to 2CCA-1 containing lysophosphatidic and phosphatidic acid in wild-type but not in fakB3 mutant bacteria. This suggests that 2CCA-1 is metabolized as a fatty acid via FakB3 and utilized as a phospholipid building block, leading to accumulation of toxic phospholipid species. Analysis of FabT-mediated fakB3 expression elucidates how the pneumococcus could ensure membrane homeostasis and concurrent economic use of host-derived fatty acids.IMPORTANCE Fatty acid biosynthesis is an attractive antibiotic target, as it affects the supply of membrane phospholipid building blocks. In Streptococcus pneumoniae, it is not sufficient to target only the endogenous fatty acid synthesis machinery, as uptake of host fatty acids may bypass this inhibition. Here, we describe a small-molecule compound, 2CCA-1, with potent bactericidal activity that upon interactions with the fatty acid binding protein FakB3, which is present in a limited number of Gram-positive species, becomes metabolized and incorporated as a toxic phospholipid species. Resistance to 2CCA-1 developed specifically in fakB3 and the regulatory gene fabT These mutants reveal a regulatory connection between the extracellular polyunsaturated fatty acid metabolism and endogenous fatty acid synthesis in S. pneumoniae, which could ensure balance between efficient scavenging of host polyunsaturated fatty acids and membrane homeostasis. The data might be useful in the identification of narrow-spectrum treatment strategies to selectively target members of the Lactobacillales such as S. pneumoniae.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carboxylic Acids/pharmacology , Fatty Acids, Unsaturated/biosynthesis , Fatty Acids/pharmacology , Streptococcus pneumoniae/drug effects , Anti-Bacterial Agents/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriolysis/drug effects , Biosynthetic Pathways/drug effects , Carboxylic Acids/chemistry , Drug Resistance, Bacterial , Fatty Acids/chemistry , Gene Expression Regulation, Bacterial , Mutation , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Streptococcus pneumoniae/cytology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism
8.
Mol Microbiol ; 113(3): 650-658, 2020 03.
Article in English | MEDLINE | ID: mdl-32185835

ABSTRACT

The Gram-positive bacterium Streptococcus pneumoniae, the pneumococcus, is an important commensal resident of the human nasopharynx. Carriage is usually asymptomatic, however, S. pneumoniae can become invasive and spread from the upper respiratory tract to the lungs causing pneumonia, and to other organs to cause severe diseases such as bacteremia and meningitis. Several pneumococcal proteins important for its disease-causing capability have been described and many are expressed on the bacterial surface. The surface located pneumococcal type-1 pilus has been associated with virulence and the inflammatory response, and it is present in 20%-30% of clinical isolates. Its tip protein RrgA has been shown to be a major adhesin to human cells and to promote invasion through the blood-brain barrier. In this review we discuss recent findings of the impact of RrgA on bacterial colonization of the upper respiratory tract and on pneumococcal virulence, and use epidemiological data and genome-mining to suggest trade-off mechanisms potentially explaining the rather low prevalence of pilus-1 expressing pneumococci in humans.


Subject(s)
Fimbriae Proteins/metabolism , Streptococcus pneumoniae/metabolism , Virulence Factors/metabolism , Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Fimbriae Proteins/physiology , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/physiology , Protein Binding , Streptococcus pneumoniae/pathogenicity , Virulence/genetics , Virulence Factors/physiology
9.
mBio ; 10(3)2019 06 25.
Article in English | MEDLINE | ID: mdl-31239383

ABSTRACT

Streptococcus pseudopneumoniae is a close relative of the major human pathogen S. pneumoniae It is increasingly associated with lower-respiratory-tract infections (LRTI) and a high prevalence of antimicrobial resistance (AMR). S. pseudopneumoniae is difficult to identify using traditional typing methods due to similarities with S. pneumoniae and other members of the mitis group (SMG). Using whole-genome sequencing of LRTI isolates and a comparative genomic approach, we found that a large number of pneumococcal virulence and colonization genes are present in the core S. pseudopneumoniae genome. We also reveal an impressive number of novel surface-exposed proteins encoded by the genome of this species. In addition, we propose a new and entirely specific molecular marker useful for the identification of S. pseudopneumoniae Phylogenetic analyses of S. pseudopneumoniae show that specific clades are associated with allelic variants of core proteins. Resistance to tetracycline and macrolides, the two most common types of resistance, were found to be encoded by Tn916-like integrating conjugative elements and Mega-2. Overall, we found a tight association of genotypic determinants of AMR and phenotypic AMR with a specific lineage of S. pseudopneumoniae Taken together, our results shed light on the distribution in S. pseudopneumoniae of genes known to be important during invasive disease and colonization and provide insight into features that could contribute to virulence, colonization, and adaptation.IMPORTANCES. pseudopneumoniae is an overlooked pathogen emerging as the causative agent of lower-respiratory-tract infections and associated with chronic obstructive pulmonary disease (COPD) and exacerbation of COPD. However, much remains unknown on its clinical importance and epidemiology, mainly due to the lack of specific markers to distinguish it from S. pneumoniae Here, we provide a new molecular marker entirely specific for S. pseudopneumoniae and offer a comprehensive view of the virulence and colonization genes found in this species. Finally, our results pave the way for further studies aiming at understanding the pathogenesis and epidemiology of S. pseudopneumoniae.


Subject(s)
Genome, Bacterial , Phylogeny , Streptococcus/genetics , Streptococcus/pathogenicity , Anti-Bacterial Agents/pharmacology , Genomics , Genotype , Humans , Respiratory Tract Infections/microbiology , Sequence Analysis, DNA , Streptococcal Infections/microbiology , Streptococcus/drug effects , Virulence/genetics , Virulence Factors/genetics , Whole Genome Sequencing
10.
Microbiol Spectr ; 7(1)2019 01.
Article in English | MEDLINE | ID: mdl-30737914

ABSTRACT

Type IV pili (T4P) are remarkable bacterial surface appendages that carry out a range of functions. Various types of T4P have been identified in bacteria and archaea, making them almost universal structures in prokaryotes. T4P are best characterized in Gram-negative bacteria, in which pilus biogenesis and T4P-mediated functions have been studied for decades. Recent advances in microbial whole-genome sequencing have provided ample evidence for the existence of T4P also in many Gram-positive species. However, comparatively little is known, and T4P in Gram-positive bacteria are just beginning to be dissected. So far, they have mainly been studied in Clostridium and Streptococcus spp. and are involved in diverse cellular processes such as adhesion, motility, and horizontal gene transfer. Here we summarize the current understanding of T4P in Gram-positive species and their functions, with particular focus on the type IV competence pilus produced by the human pathogen Streptococcus pneumoniae and its role in natural transformation.


Subject(s)
Bacterial Adhesion/genetics , Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Gene Transfer, Horizontal/genetics , Streptococcus pneumoniae/genetics , DNA Transformation Competence/genetics , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Streptococcus pneumoniae/physiology
11.
mBio ; 9(2)2018 04 10.
Article in English | MEDLINE | ID: mdl-29636428

ABSTRACT

Gram-positive bacteria, including the major respiratory pathogen Streptococcus pneumoniae, were recently shown to produce extracellular vesicles (EVs) that likely originate from the plasma membrane and are released into the extracellular environment. EVs may function as cargo for many bacterial proteins, however, their involvement in cellular processes and their interactions with the innate immune system are poorly understood. Here, EVs from pneumococci were characterized and their immunomodulatory effects investigated. Pneumococcal EVs were protruding from the bacterial surface and released into the medium as 25 to 250 nm lipid stained vesicles containing a large number of cytosolic, membrane, and surface-associated proteins. The cytosolic pore-forming toxin pneumolysin was significantly enriched in EVs compared to a total bacterial lysate but was not required for EV formation. Pneumococcal EVs were internalized into A549 lung epithelial cells and human monocyte-derived dendritic cells and induced proinflammatory cytokine responses irrespective of pneumolysin content. EVs from encapsulated pneumococci were recognized by serum proteins, resulting in C3b deposition and formation of C5b-9 membrane attack complexes as well as factor H recruitment, depending on the presence of the choline binding protein PspC. Addition of EVs to human serum decreased opsonophagocytic killing of encapsulated pneumococci. Our data suggest that EVs may act in an immunomodulatory manner by allowing delivery of vesicle-associated proteins and other macromolecules into host cells. In addition, EVs expose targets for complement factors in serum, promoting pneumococcal evasion of humoral host defense.IMPORTANCEStreptococcus pneumoniae is a major contributor to morbidity and mortality worldwide, being the major cause of milder respiratory tract infections such as otitis and sinusitis and of severe infections such as community-acquired pneumonia, with or without septicemia, and meningitis. More knowledge is needed on how pneumococci interact with the host, deliver virulence factors, and activate immune defenses. Here we show that pneumococci form extracellular vesicles that emanate from the plasma membrane and contain virulence properties, including enrichment of pneumolysin. We found that pneumococcal vesicles can be internalized into epithelial and dendritic cells and bind complement proteins, thereby promoting pneumococcal evasion of complement-mediated opsonophagocytosis. They also induce pneumolysin-independent proinflammatory responses. We suggest that these vesicles can function as a mechanism for delivery of pneumococcal proteins and other immunomodulatory components into host cells and help pneumococci to avoid complement deposition and phagocytosis-mediated killing, thereby possibly contributing to the symptoms found in pneumococcal infections.


Subject(s)
Extracellular Vesicles/immunology , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunity, Innate/drug effects , Immunologic Factors/metabolism , Streptococcus pneumoniae/immunology , Blood Bactericidal Activity , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Endocytosis , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Vesicles/metabolism , Humans , Immune Evasion , Streptococcus pneumoniae/metabolism
12.
J Clin Invest ; 126(8): 2821-6, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27348589

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is the primary cause of bacterial meningitis. Pneumococcal bacteria penetrates the blood-brain barrier (BBB), but the bacterial factors that enable this process are not known. Here, we determined that expression of pneumococcal pilus-1, which includes the pilus adhesin RrgA, promotes bacterial penetration through the BBB in a mouse model. S. pneumoniae that colonized the respiratory epithelium and grew in the bloodstream were chains of variable lengths; however, the pneumococci that entered the brain were division-competent, spherical, single cocci that expressed adhesive RrgA-containing pili. The cell division protein DivIVA, which is required for an ovoid shape, was localized at the poles and septum of pneumococcal chains of ovoid, nonseparated bacteria, but was absent in spherical, single cocci. In the bloodstream, a small percentage of pneumococci appeared as piliated, RrgA-expressing, DivIVA-negative single cocci, suggesting that only a minority of S. pneumoniae are poised to cross the BBB. Together, our data indicate that small bacterial cell size, which is signified by the absence of DivIVA, and the presence of an adhesive RrgA-containing pilus-1 mediate pneumococcal passage from the bloodstream through the BBB into the brain to cause lethal meningitis.


Subject(s)
Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Meningitis, Pneumococcal/microbiology , Streptococcus pneumoniae , Virulence Factors/metabolism , Adhesins, Bacterial , Animals , Bacterial Adhesion , Blood-Brain Barrier , Brain/microbiology , Cell Wall , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL
13.
Eur Respir J ; 47(4): 1208-18, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26797033

ABSTRACT

The effects of pneumococcal conjugated vaccines (PCVs) need to be investigated. In Stockholm County, Sweden, PCV7 was introduced in the childhood immunisation programme in 2007 and changed to PCV13 in 2010.Over 90% of all invasive isolates during 2005-2014 (n=2336) and carriage isolates, 260 before and 647 after vaccine introduction, were characterised by serotyping, molecular typing and antibiotic susceptibility, and serotype diversity was calculated. Clinical information was collected for children and adults with invasive pneumococcal disease (IPD).The IPD incidence decreased post-PCV7, but not post-PCV13, in vaccinated children. Beneficial herd effects were seen in older children and adults, but not in the elderly. The herd protection was more pronounced post-PCV7 than post-PCV13. PCV7 serotypes decreased. IPD caused by PCV13 serotypes 3 and 19A increased post-PCV7. Post-PCV13, serotypes 6A and 19A, but not serotype 3, decreased. The serotype distribution changed in carriage and IPD to nonvaccine types, also in nonvaccinated populations. Expansion of non-PCV13 serotypes was largest following PCV13 introduction. Serotype diversity increased and nonvaccine clones emerged, such as CC433 (serotype 22F) in IPD and CC62 (serotype 11A) in carriage. In young children, meningitis, septicaemia and severe rhinosinusitis, but not bacteraemic pneumonia, decreased.Pneumococcal vaccination leads to expansion of new or minor serotypes/clones, also in nonvaccinated populations.


Subject(s)
Heptavalent Pneumococcal Conjugate Vaccine/therapeutic use , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/therapeutic use , Streptococcus pneumoniae/isolation & purification , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Humans , Immunity, Herd , Incidence , Infant , Infant, Newborn , Microbial Sensitivity Tests , Middle Aged , Molecular Typing , Pneumococcal Infections/epidemiology , Serotyping , Streptococcus pneumoniae/classification , Sweden , Vaccines, Conjugate/therapeutic use , Young Adult
14.
Science ; 335(6072): 1103-6, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22383849

ABSTRACT

Bacteria adapt to environmental stimuli by adjusting their transcriptomes in a complex manner, the full potential of which has yet to be established for any individual bacterial species. Here, we report the transcriptomes of Bacillus subtilis exposed to a wide range of environmental and nutritional conditions that the organism might encounter in nature. We comprehensively mapped transcription units (TUs) and grouped 2935 promoters into regulons controlled by various RNA polymerase sigma factors, accounting for ~66% of the observed variance in transcriptional activity. This global classification of promoters and detailed description of TUs revealed that a large proportion of the detected antisense RNAs arose from potentially spurious transcription initiation by alternative sigma factors and from imperfect control of transcription termination.


Subject(s)
Bacillus subtilis/genetics , Bacillus subtilis/physiology , Gene Expression Regulation, Bacterial , Promoter Regions, Genetic , Transcription, Genetic , Transcriptome , Adaptation, Physiological , Algorithms , Binding Sites , Gene Expression Profiling , Gene Regulatory Networks , Oligonucleotide Array Sequence Analysis , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulon , Sigma Factor/metabolism , Terminator Regions, Genetic
15.
Microbiology (Reading) ; 158(Pt 3): 696-707, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22174379

ABSTRACT

The structure of the SigB-dependent general stress regulon of Bacillus subtilis has previously been characterized by proteomics approaches as well as DNA array-based expression studies. However, comparing the SigB targets published in three previous major transcriptional profiling studies it is obvious that although each of them identified well above 100 target genes, only 67 were identified in all three studies. These substantial differences can likely be attributed to the different strains, growth conditions, microarray platforms and experimental setups used in the studies. In order to gain a better understanding of the structure of this important regulon, a targeted DNA microarray analysis covering most of the known SigB-inducing conditions was performed, and the changes in expression kinetics of 252 potential members of the SigB regulon and appropriate control genes were recorded. Transcriptional data for the B. subtilis wild-type strain 168 and its isogenic sigB mutant BSM29 were analysed using random forest, a machine learning algorithm, by incorporating the knowledge from previous studies. This analysis revealed a strictly SigB-dependent expression pattern for 166 genes following ethanol, butanol, osmotic and oxidative stress, low-temperature growth and heat shock, as well as limitation of oxygen or glucose. Kinetic analysis of the data for the wild-type strain identified 30 additional members of the SigB regulon, which were also subject to control by additional transcriptional regulators, thus displaying atypical SigB-independent induction patterns in the mutant strain under some of the conditions tested. For 19 of these 30 SigB regulon members, published reports support control by secondary regulators along with SigB. Thus, this microarray-based study assigns a total of 196 genes to the SigB-dependent general stress regulon of B. subtilis.


Subject(s)
Bacillus subtilis/genetics , Bacterial Proteins/metabolism , Gene Expression Profiling , Regulon , Sigma Factor/metabolism , Stress, Physiological , Bacillus subtilis/physiology , Bacterial Proteins/genetics , Gene Deletion , Microarray Analysis , Sigma Factor/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...