Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.442
Filtrar
1.
Int J Mol Sci ; 25(17)2024 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-39273173

RESUMO

Escherichia coli O157:H7 (E. coli O157) is known for causing severe foodborne illnesses such as hemorrhagic colitis and hemolytic uremic syndrome. Although E. coli O157 is typically regarded as an extracellular pathogen and a weak biofilm producer, some E. coli O157 strains, including a clinical strain ATCC 43895, exhibit a notable ability to invade bovine crypt cells and other epithelial cells, as well as to form robust biofilm. This invasive strain persists in the bovine host significantly longer than non-invasive strains. Various surface-associated factors, including lipopolysaccharides (LPS), flagella, and other adhesins, likely contribute to this enhanced invasiveness and biofilm formation. In this study, we constructed a series of LPS-core deletion mutations (waaI, waaG, waaF, and waaC) in E. coli O157 ATCC 43895, resulting in stepwise truncations of the LPS. This approach enabled us to investigate the effects on the biosynthesis of key surface factors, such as flagella and curli, and the ability of this invasive strain to invade host cells. We confirmed the LPS structure and found that all LPS-core mutants failed to form biofilms, highlighting the crucial role of core oligosaccharides in biofilm formation. Additionally, the LPS inner-core mutants ΔwaaF and ΔwaaC lost the ability to produce flagella and curli. Furthermore, these inner-core mutants exhibited a dramatic reduction in adherence to and invasion of epithelial cells (MAC-T), showing an approximately 100-fold decrease in cell invasion compared with the outer-core mutants (waaI and waaG) and the wild type. These findings underscore the critical role of LPS-core truncation in impairing flagella and curli biosynthesis, thereby reducing the invasion capability of E. coli O157 ATCC 43895.


Assuntos
Biofilmes , Escherichia coli O157 , Flagelos , Lipopolissacarídeos , Flagelos/metabolismo , Flagelos/genética , Lipopolissacarídeos/biossíntese , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Escherichia coli O157/fisiologia , Biofilmes/crescimento & desenvolvimento , Animais , Bovinos , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Aderência Bacteriana , Células Epiteliais/microbiologia , Células Epiteliais/metabolismo
2.
Braz J Microbiol ; 55(3): 2727-2738, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39222218

RESUMO

Due to the increasing occurrence of drug resistant urinary tract infections (UTI) among children, there is a need to investigate alternative effective treatment protocols such as nanoparticles. Flagella and fimbriae are primary factors contributing the virulence of urinary tract infecting bacteria. The aim of this study was to assess the antibacterial effects of zinc oxide nanoparticles which have been synthesized using both chemical and green methods on multi-drug resistant (MDR) uropathogenic bacteria encoding fli and fim genes and investigating their binding ability to bacterial appendage proteins. A total of 30 urine culture samples were collected from children under 2 years old diagnosed with urinary tract infection. The isolates underwent antibiotic suseptibility assessment and the isolates demonstrating MDR were subjected to molecular amplification of fimG (fimbrial) and fliD and fliT (flagellal) genes. The confirmation of cellular appendages was achieved through silver nitrate staining. The antibacterial efficacy of the synthetized nanoparticles was assessed using the micro and macrodilution methods. The successful binding of nanoparticles to bacterial appendage proteins was confirmed through mobility shift and membrane filter assays. The dimensions of chemically synthesized ZnO nanoparticles and green nanoparticles were measured at 30 nm and 85 nm, respectively, with the exhibition of hexagonal geometries. The nanoparticles synthesized through chemical and green methods exhibited minimum inhibitory concentrations (MIC) of 0.0062-0.025 g/L and 0.3 g/L, respectively. The ability of ZnO nanoparticles to bind bacterial appendage proteins and to combat MDR uropathogenic bacteria are promising for new treatment protocols against UTI in children in future.


Assuntos
Antibacterianos , Farmacorresistência Bacteriana Múltipla , Flagelos , Infecções Urinárias , Óxido de Zinco , Óxido de Zinco/farmacologia , Óxido de Zinco/química , Óxido de Zinco/metabolismo , Antibacterianos/farmacologia , Humanos , Infecções Urinárias/microbiologia , Infecções Urinárias/tratamento farmacológico , Flagelos/efeitos dos fármacos , Flagelos/genética , Flagelos/metabolismo , Testes de Sensibilidade Microbiana , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/metabolismo , Fímbrias Bacterianas/efeitos dos fármacos , Nanopartículas/química , Lactente , Nanopartículas Metálicas/química
3.
ISME J ; 18(1)2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-39113613

RESUMO

Biofilms aid bacterial adhesion to surfaces via direct and indirect mechanisms, and formation of biofilms is considered as an important strategy for adaptation and survival in suboptimal environmental conditions. However, the molecular underpinnings of biofilm formation in subsurface sediment/groundwater ecosystems where microorganisms often experience fluctuations in nutrient input, pH, and nitrate or metal concentrations are underexplored. We examined biofilm formation under different nutrient, pH, metal, and nitrate regimens of 16 Rhodanobacter strains isolated from subsurface groundwater wells spanning diverse levels of pH (3.5 to 5) and nitrates (13.7 to 146 mM). Eight Rhodanobacter strains demonstrated significant biofilm growth under low pH, suggesting adaptations for survival and growth at low pH. Biofilms were intensified under aluminum stress, particularly in strains possessing fewer genetic traits associated with biofilm formation, findings warranting further investigation. Through random barcode transposon-site sequencing (RB-TnSeq), proteomics, use of specific mutants, and transmission electron microscopy analysis, we discovered flagellar loss under aluminum stress, indicating a potential relationship between motility, metal tolerance, and biofilm growth. Comparative genomic analyses revealed the absence of flagella and chemotaxis genes and the presence of a putative type VI secretion system in the highly biofilm-forming strain FW021-MT20. In this study we identified genetic determinants associated with biofilm growth under metal stress in a predominant environmental genus, Rhodanobacter, and identified traits aiding survival and adaptation to contaminated subsurface environments.


Assuntos
Adaptação Fisiológica , Alumínio , Biofilmes , Flagelos , Estresse Fisiológico , Biofilmes/crescimento & desenvolvimento , Flagelos/genética , Flagelos/fisiologia , Alumínio/toxicidade , Concentração de Íons de Hidrogênio , Nitratos/metabolismo , Água Subterrânea/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
4.
Fish Shellfish Immunol ; 151: 109752, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38977112

RESUMO

Pseudomonas plecoglossicida is a vital pathogen that poses a substantial risk to aquaculture. Small RNAs (sRNAs) are non-coding regulatory molecules capable of sensing environmental changes and modulating virulence-associated signaling pathways, such as the assembly of flagella. However, the relevant researches on P. plecoglossicida are an urgent need. Here, we report a novel sRNA, sRNA562, which has potential to regulate the post-transcriptional of fliP, a key component of the lateral flagellar type III secretion system. In this study, the effects of sRNA562 on the virulence of P. plecoglossicida and its role in regulating the pathogenic process were investigated through the use of a constructed sRNA562 deletion strain. The deletion of sRNA562 resulted in an up-regulation of fliP in P. plecoglossicida, and leading to increased swarming motility and enhanced the ability of biofilm formation, adhesion and chemotaxis. Subsequent artificial infection experiment demonstrated that the deletion of sRNA562 increased the virulence of P. plecoglossicida towards hybrid grouper, as evidenced by a reduction in survival rate, elevation of tissue bacterial load, and the exacerbation of histopathological damage. Further studies have found that the deletion of sRNA562 lead to an up-regulation of fliP expression during hybrid grouper infection, thereby enhancing bacterial swarming ability and ultimately heightening pathogenicity, leading to a dysregulated host response to infection, tissue damage and eventually death. Our work revealed a sRNA that exerts negative regulation on the expression of lateral flagella in P. plecoglossicida, thereby impacting its virulence. These findings provide a new perspective on the virulence regulation mechanism of P. plecoglossicida, contributing to a more comprehensive understanding in the field of pathogenicity research.


Assuntos
Doenças dos Peixes , Flagelos , Regulação Bacteriana da Expressão Gênica , Pseudomonas , Pequeno RNA não Traduzido , Pseudomonas/patogenicidade , Pseudomonas/genética , Pseudomonas/fisiologia , Virulência/genética , Animais , Doenças dos Peixes/microbiologia , Pequeno RNA não Traduzido/genética , Flagelos/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , RNA Bacteriano/genética , Sistemas de Secreção Tipo III/genética , Bass , Infecções por Pseudomonas/imunologia
5.
Mol Microbiol ; 122(2): 255-270, 2024 08.
Artigo em Inglês | MEDLINE | ID: mdl-39030901

RESUMO

The flagellar MS-ring, uniquely constituted by FliF, is essential for flagellar biogenesis and functionality in several bacteria. The aim of this study was to dissect the role of FliF in the Gram-positive and peritrichously flagellated Bacillus cereus. We demonstrate that fliF forms an operon with the upstream gene fliE. In silico analysis of B. cereus ATCC 14579 FliF identifies functional domains and amino acid residues that are essential for protein functioning. The analysis of a ΔfliF mutant of B. cereus, constructed in this study using an in frame markerless gene replacement method, reveals that the mutant is unexpectedly able to assemble flagella, although in reduced amounts compared to the parental strain. Nevertheless, motility is completely abolished by fliF deletion. FliF deprivation causes the production of submerged biofilms and affects the ability of B. cereus to adhere to gastrointestinal mucins. We additionally show that the fliF deletion does not compromise the secretion of the three components of hemolysin BL, a toxin secreted through the flagellar type III secretion system. Overall, our findings highlight the important role of B. cereus FliF in flagella-related functions, being the protein required for complete flagellation, motility, mucin adhesion, and pellicle biofilms.


Assuntos
Bacillus cereus , Proteínas de Bactérias , Biofilmes , Flagelos , Óperon , Bacillus cereus/metabolismo , Bacillus cereus/genética , Flagelos/metabolismo , Flagelos/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Biofilmes/crescimento & desenvolvimento , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/genética , Aderência Bacteriana , Regulação Bacteriana da Expressão Gênica , Deleção de Genes , Proteínas de Membrana
6.
J Assist Reprod Genet ; 41(9): 2271-2278, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38963606

RESUMO

PURPOSE: To identify novel variants in ACTL9 and new phenotypes responsible for male infertility. METHODS: Genomic DNA was extracted from peripheral blood samples for whole-exome sequencing (WES). Computer-assisted sperm analysis (CASA) was used to test the motility of spermatozoa. The ultrastructure of flagella and the mitochondrial sheath were assessed by scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Immunostaining was used to validate the localization and expression of ACTL9 and ACTL7A. An Actl9-mutated mouse model was used to validate the phenotypes by CASA and TEM. RESULTS: We identified novel homozygous variants in ACTL9 in two independent Chinese families. Spermatozoa with ACTL9 mutations showed decreased CASA parameters and a higher proportion of spermatozoa with abnormal morphology, exhibiting coiled flagella and a thickened midpiece. The spermatozoa were characterized by chaotic or irregular '9+2' structures and irregular mitochondrial sheath arrangements in the flagellum. Actl9 knock-in mice also showed abnormal CASA parameters and irregular '9+2' structures in flagella. CONCLUSIONS: Our study expands the mutation spectrum and phenotypic spectrum of ACTL9.


Assuntos
Flagelos , Homozigoto , Infertilidade Masculina , Mitocôndrias , Mutação , Motilidade dos Espermatozoides , Cauda do Espermatozoide , Espermatozoides , Masculino , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Humanos , Camundongos , Espermatozoides/patologia , Espermatozoides/ultraestrutura , Espermatozoides/metabolismo , Animais , Mitocôndrias/genética , Mitocôndrias/ultraestrutura , Mitocôndrias/patologia , Mitocôndrias/metabolismo , Mutação/genética , Cauda do Espermatozoide/patologia , Cauda do Espermatozoide/metabolismo , Cauda do Espermatozoide/ultraestrutura , Flagelos/genética , Flagelos/ultraestrutura , Flagelos/metabolismo , Motilidade dos Espermatozoides/genética , Sequenciamento do Exoma , Linhagem , Adulto , Análise do Sêmen
8.
Int J Mol Sci ; 25(13)2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-39000482

RESUMO

Plesiomonas shigelloides, a Gram-negative bacillus, is the only member of the Enterobacteriaceae family able to produce polar and lateral flagella and cause gastrointestinal and extraintestinal illnesses in humans. The flagellar transcriptional hierarchy of P. shigelloides is currently unknown. In this study, we identified FlaK, FlaM, FliA, and FliAL as the four regulators responsible for polar and lateral flagellar regulation in P. shigelloides. To determine the flagellar transcription hierarchy of P. shigelloides, the transcriptomes of the WT and ΔflaK, ΔflaM, ΔfliA, and ΔfliAL were carried out for comparison in this study. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) and luminescence screening assays were used to validate the RNA-seq results, and the Electrophoretic Mobility Shift Assay (EMSA) results revealed that FlaK can directly bind to the promoters of fliK, fliE, flhA, and cheY, while the FlaM protein can bind directly to the promoters of flgO, flgT, and flgA. Meanwhile, we also observed type VI secretion system (T6SS) and type II secretion system 2 (T2SS-2) genes downregulated in the transcriptome profiles, and the killing assay revealed lower killing abilities for ΔflaK, ΔflaM, ΔfliA, and ΔfliAL compared to the WT, indicating that there was a cross-talk between the flagellar hierarchy system and bacterial secretion system. Invasion assays also showed that ΔflaK, ΔflaM, ΔfliA, and ΔfliAL were less effective in infecting Caco-2 cells than the WT. Additionally, we also found that the loss of flagellar regulators causes the differential expression of some of the physiological metabolic genes of P. shigelloides. Overall, this study aims to reveal the transcriptional hierarchy that controls flagellar gene expression in P. shigelloides, as well as the cross-talk between motility, virulence, and physiological and metabolic activity, laying the groundwork for future research into P. shigelloides' coordinated survival in the natural environment and the mechanisms that infect the host.


Assuntos
Proteínas de Bactérias , Flagelos , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Plesiomonas , Flagelos/metabolismo , Flagelos/genética , Plesiomonas/genética , Plesiomonas/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Transcriptoma , Regiões Promotoras Genéticas , Sistemas de Secreção Bacterianos/genética , Sistemas de Secreção Bacterianos/metabolismo , Transcrição Gênica , Humanos
9.
BMC Microbiol ; 24(1): 234, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38951769

RESUMO

BACKGROUND: Klebsiella aerogenes is an opportunistic pathogen that causes a wide variety of infections. Due to the rising problem of antibiotic resistance, novel antibiotics and strategies to combat bacterial infections are needed. Host-specific bacteriophages are natural enemies of bacteria and can be used in phage therapy as an alternative form of treatment against bacterial infections. Jumbo phages are defined as phages with genomes larger than 200 kb. Relatively few studies have been done on jumbo phages compared to smaller phages. RESULTS: A novel phage, fENko-Kae01, was isolated from a commercial phage cocktail. Genomic analysis revealed that fENko-Kae01 is a lytic jumbo phage with a 360 kb genome encoding 578 predicted genes. No highly similar phage genomes were identified and fENko-Kae01 may be a completely new genus representative. No known genes associated with lysogenic life cycle, bacterial virulence, or antibiotic resistance were identified. The phage had myovirus morphology and a narrow host range. Phage resistant bacterial mutants emerged under phage selection. Whole genome sequencing revealed that the biogenesis of the flagellum was affected in four mutants and the lack of functional flagellum was confirmed in motility assays. Furthermore, phage fENKo-Kae01 failed to adsorb on the non-motile mutants indicating that the bacterial flagellum is the phage-binding receptor. CONCLUSIONS: fENko-Kae01 is a novel jumbo bacteriophage that is considered safe for phage therapy. fENko-Kae01 uses the flagellum as the phage-binding receptor and may represent a completely novel genus.


Assuntos
Bacteriófagos , Enterobacter aerogenes , Flagelos , Genoma Viral , Especificidade de Hospedeiro , Bacteriófagos/genética , Bacteriófagos/classificação , Bacteriófagos/isolamento & purificação , Bacteriófagos/fisiologia , Flagelos/virologia , Flagelos/genética , Enterobacter aerogenes/virologia , Enterobacter aerogenes/genética , Sequenciamento Completo do Genoma , Myoviridae/genética , Myoviridae/isolamento & purificação , Myoviridae/classificação , Myoviridae/fisiologia
10.
Nat Commun ; 15(1): 5921, 2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39004688

RESUMO

The bacterial flagellum, which facilitates motility, is composed of ~20 structural proteins organized into a long extracellular filament connected to a cytoplasmic rotor-stator complex via a periplasmic rod. Flagellum assembly is regulated by multiple checkpoints that ensure an ordered gene expression pattern coupled to the assembly of the various building blocks. Here, we use epifluorescence, super-resolution, and transmission electron microscopy to show that the absence of a periplasmic protein (FlhE) prevents proper flagellar morphogenesis and results in the formation of periplasmic flagella in Salmonella enterica. The periplasmic flagella disrupt cell wall synthesis, leading to a loss of normal cell morphology resulting in cell lysis. We propose that FlhE functions as a periplasmic chaperone to control assembly of the periplasmic rod, thus preventing formation of periplasmic flagella.


Assuntos
Proteínas de Bactérias , Flagelos , Chaperonas Moleculares , Periplasma , Flagelos/metabolismo , Flagelos/ultraestrutura , Flagelos/genética , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/genética , Periplasma/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Salmonella enterica/metabolismo , Salmonella enterica/genética , Microscopia Eletrônica de Transmissão , Proteínas Periplásmicas/metabolismo , Proteínas Periplásmicas/genética , Regulação Bacteriana da Expressão Gênica
11.
mBio ; 15(8): e0071524, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39037271

RESUMO

The bacterial predator Bdellovibrio bacteriovorus is considered to be obligatorily prey (host)-dependent (H-D), and thus unable to form biofilms. However, spontaneous host-independent (H-I) variants grow axenically and can form robust biofilms. A screen of 350 H-I mutants revealed that single mutations in stator genes fliL or motA were sufficient to generate flagellar motility-defective H-I strains able to adhere to surfaces but unable to develop biofilms. The variants showed large transcriptional shifts in genes related to flagella, prey-invasion, and cyclic-di-GMP (CdG), as well as large changes in CdG cellular concentration relative to the H-D parent. The introduction of the parental fliL allele resulted in a full reversion to the H-D phenotype, but we propose that specific interactions between stator proteins prevented functional complementation by fliL paralogs. In contrast, specific mutations in a pilus-associated protein (Bd0108) mutant background were necessary for biofilm formation, including secretion of extracellular DNA (eDNA), proteins, and polysaccharides matrix components. Remarkably, fliL disruption strongly reduced biofilm development. All H-I variants grew similarly without prey, showed a strain-specific reduction in predatory ability in prey suspensions, but maintained similar high efficiency in prey biofilms. Population-wide allele sequencing suggested additional routes to host independence. Thus, stator and invasion pole-dependent signaling control the H-D and the H-I biofilm-forming phenotypes, with single mutations overriding prey requirements, and enabling shifts from obligate to facultative predation, with potential consequences on community dynamics. Our findings on the facility and variety of changes leading to facultative predation also challenge the concept of Bdellovibrio and like organisms being obligate predators. IMPORTANCE: The ability of bacteria to form biofilms is a central research theme in biology, medicine, and the environment. We show that cultures of the obligate (host-dependent) "solitary" predatory bacterium Bdellovibrio bacteriovorus, which cannot replicate without prey, can use various genetic routes to spontaneously yield host-independent (H-I) variants that grow axenically (as a single species, in the absence of prey) and exhibit various surface attachment phenotypes, including biofilm formation. These routes include single mutations in flagellar stator genes that affect biofilm formation, provoke motor instability and large motility defects, and disrupt cyclic-di-GMP intracellular signaling. H-I strains also exhibit reduced predatory efficiency in suspension but high efficiency in prey biofilms. These changes override the requirements for prey, enabling a shift from obligate to facultative predation, with potential consequences on community dynamics.


Assuntos
Proteínas de Bactérias , Bdellovibrio bacteriovorus , Biofilmes , Flagelos , Biofilmes/crescimento & desenvolvimento , Flagelos/genética , Flagelos/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bdellovibrio bacteriovorus/genética , Bdellovibrio bacteriovorus/fisiologia , Mutação , Regulação Bacteriana da Expressão Gênica , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo
12.
Lett Appl Microbiol ; 77(7)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38906839

RESUMO

Vibrio parahaemolyticus utilizes a polar flagellum for swimming in liquids and employs multiple lateral flagella to swarm on surfaces and in viscous environments. The VPA0961 protein is an LysR family transcriptional regulator that can regulate the swimming and swarming motility of V. parahaemolyticus, but the detailed regulatory mechanisms are not yet fully understood. Herein, we designated the protein as AcsS, which stands for activator of swimming and swarming motility. Our data provided evidence that deleting the acsS gene significantly reduced both swimming and swarming motility of V. parahaemolyticus. Furthermore, AcsS was found to activate the expression of both polar (flgA, flgM, flgB, and flgK) and lateral (motY, fliM, lafA, and fliD) flagellar genes. Overexpression of AcsS in Escherichia coli induced the expression of flgA, motY, and lafA, but did not affect the expression of flgB, flgK, flgM, fliM, and fliD. Interestingly, His-tagged AcsS did not bind to the upstream DNA regions of all the tested genes, suggesting indirect regulation. In conclusion, AcsS positively regulated the swimming and swarming motility of V. parahaemolyticus by activating the transcription of polar and lateral flagellar genes. This work enriched our understanding of the gene expression regulation within the dual flagellar systems of V. parahaemolyticus.


Assuntos
Proteínas de Bactérias , Flagelos , Regulação Bacteriana da Expressão Gênica , Fatores de Transcrição , Vibrio parahaemolyticus , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/fisiologia , Flagelos/genética , Flagelos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo
13.
Nat Commun ; 15(1): 5240, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38897989

RESUMO

Like for many bacteria, flagella are crucial for Campylobacter jejuni motility and virulence. Biogenesis of the flagellar machinery requires hierarchical transcription of early, middle (RpoN-dependent), and late (FliA-dependent) genes. However, little is known about post-transcriptional regulation of flagellar biogenesis by small RNAs (sRNAs). Here, we characterized two sRNAs with opposing effects on C. jejuni filament assembly and motility. We demonstrate that CJnc230 sRNA (FlmE), encoded downstream of the flagellar hook protein, is processed from the RpoN-dependent flgE mRNA by RNase III, RNase Y, and PNPase. We identify mRNAs encoding a flagella-interaction regulator and the anti-sigma factor FlgM as direct targets of CJnc230 repression. CJnc230 overexpression upregulates late genes, including the flagellin flaA, culminating in longer flagella and increased motility. In contrast, overexpression of the FliA-dependent sRNA CJnc170 (FlmR) reduces flagellar length and motility. Overall, our study demonstrates how the interplay of two sRNAs post-transcriptionally fine-tunes flagellar biogenesis through balancing of the hierarchically-expressed components.


Assuntos
Proteínas de Bactérias , Campylobacter jejuni , Flagelos , Regulação Bacteriana da Expressão Gênica , RNA Bacteriano , Pequeno RNA não Traduzido , Campylobacter jejuni/genética , Campylobacter jejuni/metabolismo , Flagelos/genética , Flagelos/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , Pequeno RNA não Traduzido/genética , Pequeno RNA não Traduzido/metabolismo , Flagelina/metabolismo , Flagelina/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Ribonuclease III/metabolismo , Ribonuclease III/genética
14.
J Cell Biol ; 223(9)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-38829962

RESUMO

Two sets of motor proteins underpin motile cilia/flagella function. The axoneme-associated inner and outer dynein arms drive sliding of adjacent axoneme microtubule doublets to periodically bend the flagellum for beating, while intraflagellar transport (IFT) kinesins and dyneins carry IFT trains bidirectionally along the axoneme. Despite assembling motile cilia and flagella, IFT train speeds have only previously been quantified in immobilized flagella-mechanical immobilization or genetic paralysis. This has limited investigation of the interaction between IFT and flagellar beating. Here, in uniflagellate Leishmania parasites, we use high-frequency, dual-color fluorescence microscopy to visualize IFT train movement in beating flagella. We discovered that adhesion of flagella to a microscope slide is detrimental, reducing IFT train speed and increasing train stalling. In flagella free to move, IFT train speed is not strongly dependent on flagella beat type; however, permanent disruption of flagella beating by deletion of genes necessary for formation or regulation of beating showed an inverse correlation of beat frequency and IFT train speed.


Assuntos
Flagelos , Leishmania , Microtúbulos , Axonema/metabolismo , Axonema/genética , Transporte Biológico , Cílios/metabolismo , Cílios/genética , Dineínas/metabolismo , Dineínas/genética , Flagelos/metabolismo , Flagelos/genética , Cinesinas/metabolismo , Cinesinas/genética , Leishmania/citologia , Leishmania/genética , Leishmania/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Microtúbulos/metabolismo
15.
mBio ; 15(7): e0104824, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-38874412

RESUMO

More than half of women will experience a urinary tract infection (UTI) with most cases caused by uropathogenic Escherichia coli (UPEC). Bacterial swimming motility enhances UPEC pathogenicity, resulting in more severe disease outcomes including kidney infection. Surprisingly, the connection between motility and iron limitation is mostly unexplored despite the lack of free iron available in the host. We sought to investigate a potential connection between iron restriction and regulation of motility in UPEC. We cultured E. coli CFT073, a prototypical UPEC strain, under iron limitation and observed that CFT073 had elevated fliC (flagella) promoter activity, and this iron-specific response was repressed by the addition of exogenous iron. We confirmed increased flagellar expression in CFT073 by measuring fliC transcript, FliC protein, and surface-expressed flagella under iron-limited conditions. Interestingly, known motility regulator flhDC did not have altered transcription under these conditions. To define the regulatory mechanism of this response, we constructed single knockouts of eight master regulators and found the iron-regulated response was lost in crp, arcA, and fis mutants. Thus, we focused on the five genes regulated by all three regulators. Of the five genes knocked out, the iron-regulated motility response was most strongly dysregulated in the lpdA mutant, which also resulted in significantly lowered fitness in the murine model of ascending UTI, both against the WT and a non-motile fliC mutant. Collectively, we demonstrated that iron-mediated motility in CFT073 is partially regulated by lpdA, which contributes to the understanding of how uropathogens differentially regulate motility mechanisms in the iron-restricted host. IMPORTANCE: Urinary tract infections (UTIs) are ubiquitous and responsible for over five billion dollars in associated health care costs annually. Both iron acquisition and motility are highly studied virulence factors associated with uropathogenic Escherichia coli (UPEC), the main causative agent of uncomplicated UTI. This work is innovative by providing mechanistic insight into the synergistic relationship between these two critical virulence properties. Here, we demonstrate that iron limitation has pleiotropic effects with consequences that extend beyond metabolism and impact other virulence mechanisms. Indeed, targeting iron acquisition as a therapy may lead to an undesirable enhancement of UPEC pathogenesis through increased motility. It is vital to understand the full breadth of UPEC pathogenesis to adequately respond to this common infection, especially with the increase of antibiotic-resistant pathogens.


Assuntos
Infecções por Escherichia coli , Proteínas de Escherichia coli , Regulação Bacteriana da Expressão Gênica , Infecções Urinárias , Escherichia coli Uropatogênica , Animais , Feminino , Camundongos , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Flagelos/genética , Flagelos/metabolismo , Flagelina , Ferro/metabolismo , Locomoção , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/efeitos dos fármacos , Escherichia coli Uropatogênica/genética , Escherichia coli Uropatogênica/metabolismo , Escherichia coli Uropatogênica/patogenicidade , Virulência
16.
Microb Pathog ; 193: 106752, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38880315

RESUMO

Arcobacter butzleri is a foodborne pathogen that mainly causes enteritis in humans, but the number of cases of bacteraemia has increased in recent years. However, there is still limited knowledge on the pathogenic mechanisms of this bacterium. To investigate how A. butzleri causes disease, single knockout mutants were constructed in the cadF, ABU_RS00335, ciaB, and flaAB genes, which might be involved in adhesion and invasion properties. These mutants and the isogenic wild-type (WT) were then tested for their ability to adhere and invade human Caco-2 and HT29-MTX cells. The adhesion and invasion of A. butzleri RM4018 strain was also visualized by a Leica CTR 6500 confocal microscope. The adhesion and invasion abilities of mutants lacking the invasion antigen CiaB or a functional flagellum were lower than those of the WTs. However, the extent of the decrease varied depending on the strain and/or cell line. Mutants lacking the fibronectin (FN)-binding protein CadF consistently exhibited reduced abilities, while the inactivation of the other studied FN-binding protein, ABU_RS00335, led to a reduction in only one of the two strains tested. Therefore, the ciaB and flaAB genes appear to be important for A. butzleri adhesion and invasion properties, while cadF appears to be indispensable.


Assuntos
Adesinas Bacterianas , Arcobacter , Aderência Bacteriana , Flagelos , Aderência Bacteriana/genética , Humanos , Arcobacter/genética , Células CACO-2 , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Flagelos/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Técnicas de Inativação de Genes , Células HT29 , Fibronectinas/metabolismo , Fibronectinas/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Genes Bacterianos/genética , Células Epiteliais/microbiologia , Virulência/genética
17.
Microbiol Res ; 286: 127811, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38909416

RESUMO

Clostridioides difficile infection (CDI) caused by toxigenic C. difficile is the leading cause of antimicrobial and healthcare-associated diarrhea. The pathogenicity of C. difficile relies on the synergistic effect of multiple virulence factors, including spores, flagella, type IV pili (T4P), toxins, and biofilm. Spores enable survival and transmission of C. difficile, while adhesion factors such as flagella and T4P allow C. difficile to colonize and persist in the host intestine. Subsequently, C. difficile produces the toxins TcdA and TcdB, causing pseudomembranous colitis and other C. difficile-associated diseases; adhesion factors bind to the extracellular matrix to form biofilm, allowing C. difficile to evade drug and immune system attack and cause recurrent infection. Cyclic diguanylate (c-di-GMP) is a near-ubiquitous second messenger that extensively regulates morphology, the expression of virulence factors, and multiple physiological processes in C. difficile. In this review, we summarize current knowledge of how c-di-GMP differentially regulates the expression of virulence factors and pathogenesis-related phenotypes in C. difficile. We highlight that C. difficile spore formation and expression of toxin and flagella genes are inhibited at high intracellular levels of c-di-GMP, while T4P biosynthesis, cell aggregation, and biofilm formation are induced. Recent studies have enhanced our understanding of the c-di-GMP signaling networks in C. difficile and provided insights for the development of c-di-GMP-dependent strategies against CDI.


Assuntos
Proteínas de Bactérias , Biofilmes , Clostridioides difficile , Infecções por Clostridium , GMP Cíclico , Regulação Bacteriana da Expressão Gênica , Fenótipo , Fatores de Virulência , Clostridioides difficile/patogenicidade , Clostridioides difficile/genética , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Biofilmes/crescimento & desenvolvimento , Humanos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Infecções por Clostridium/microbiologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Esporos Bacterianos/genética , Flagelos/genética , Virulência , Enterotoxinas/genética , Enterotoxinas/metabolismo , Animais
18.
J Mol Evol ; 92(4): 381-401, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38926179

RESUMO

Kinesins are eukaryotic microtubule motor proteins subdivided into conserved families with distinct functional roles. While many kinesin families are widespread in eukaryotes, each organismal lineage maintains a unique kinesin repertoire composed of many families with distinct numbers of genes. Previous genomic surveys indicated that land plant kinesin repertoires differ markedly from other eukaryotes. To determine when repertoires diverged during plant evolution, we performed robust phylogenomic analyses of kinesins in 24 representative plants, two algae, two animals, and one yeast. These analyses show that kinesin repertoires expand and contract coincident with major shifts in the biology of algae and land plants. One kinesin family and five subfamilies, each defined by unique domain architectures, emerged in the green algae. Four of those kinesin groups expanded in ancestors of modern land plants, while six other kinesin groups were lost in the ancestors of pollen-bearing plants. Expansions of different kinesin families and subfamilies occurred in moss and angiosperm lineages. Other kinesin families remained stable and did not expand throughout plant evolution. Collectively these data support a radiation of kinesin domain architectures in algae followed by differential positive and negative selection on kinesins families and subfamilies in different lineages of land plants.


Assuntos
Evolução Molecular , Flagelos , Cinesinas , Animais , Flagelos/genética , Cinesinas/genética , Filogenia , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Plantas/genética , Domínios Proteicos
19.
Microbiol Res ; 285: 127775, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38788350

RESUMO

Vibrio alginolyticus is one of the most common opportunistic pathogens in marine animals and humans. In this study, A transposon mutation library of the V. alginolyticus E110 was used to identify motility-related genes, and we found three flagellar and one capsular polysaccharide (CPS) synthesis-related genes were linked to swarming motility. Then, gene deletion and complementation further confirmed that CPS synthesis-related gene ugd is involved in the swarming motility of V. alginolyticus. Phenotype assays showed that the Δugd mutant reduced CPS production, decreased biofilm formation, impaired swimming ability, and increased cytotoxicity compared to the wild-type strain. Transcriptome analysis showed that 655 genes (15%) were upregulated and 914 genes (21%) were downregulated in the Δugd strain. KEGG pathway and heatmap analysis revealed that genes involved in two-component systems (TCSs), chemotaxis, and flagella assembly pathways were downregulated in the Δugd mutant. On the other hand, genes involved in pathways of human diseases, biosynthesis ABC transporters, and metabolism were upregulated in the Δugd mutant. The RT-qPCR further validated that ugd-regulated genes are associated with motility, biofilm formation, virulence, and TCSs. These findings imply that ugd may be an important player in the control of some physiological processes in V. alginolyticus, highlighting its potential as a target for future research and potential therapeutic interventions.


Assuntos
Cápsulas Bacterianas , Proteínas de Bactérias , Biofilmes , Flagelos , Regulação Bacteriana da Expressão Gênica , Vibrio alginolyticus , Vibrio alginolyticus/genética , Vibrio alginolyticus/fisiologia , Vibrio alginolyticus/metabolismo , Biofilmes/crescimento & desenvolvimento , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Flagelos/genética , Flagelos/metabolismo , Flagelos/fisiologia , Cápsulas Bacterianas/metabolismo , Cápsulas Bacterianas/genética , Polissacarídeos Bacterianos/biossíntese , Polissacarídeos Bacterianos/metabolismo , Polissacarídeos Bacterianos/genética , Virulência , Animais , Perfilação da Expressão Gênica , Deleção de Genes , Humanos , Vibrioses/microbiologia
20.
mBio ; 15(6): e0044024, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38700325

RESUMO

Motility promotes biofilm initiation during the early steps of this process: microbial surface association and attachment. Motility is controlled in part by chemotaxis signaling, so it seems reasonable that chemotaxis may also affect biofilm formation. There is a gap, however, in our understanding of the interactions between chemotaxis and biofilm formation, partly because most studies analyzed the phenotype of only a single chemotaxis signaling mutant, e.g., cheA. Here, we addressed the role of chemotaxis in biofilm formation using a full set of chemotaxis signaling mutants in Helicobacter pylori, a class I carcinogen that infects more than half the world's population and forms biofilms. Using mutants that lack each chemotaxis signaling protein, we found that chemotaxis signaling affected the biofilm initiation stage, but not mature biofilm formation. Surprisingly, some chemotaxis mutants elevated biofilm initiation, while others inhibited it in a manner that was not tied to chemotaxis ability or ligand input. Instead, the biofilm phenotype correlated with flagellar rotational bias. Specifically, mutants with a counterclockwise bias promoted biofilm initiation, e.g., ∆cheA, ∆cheW, or ∆cheV1; in contrast, those with a clockwise bias inhibited it, e.g., ∆cheZ, ∆chePep, or ∆cheV3. We tested this correlation using a counterclockwise bias-locked flagellum, which induced biofilm formation independent of the chemotaxis system. These CCW flagella, however, were not sufficient to induce biofilm formation, suggesting there are downstream players. Overall, our work highlights the new finding that flagellar rotational direction promotes biofilm initiation, with the chemotaxis signaling system operating as one mechanism to control flagellar rotation. IMPORTANCE: Chemotaxis signaling systems have been reported to contribute to biofilm formation in many bacteria; however, how they regulate biofilm formation remains largely unknown. Chemotaxis systems are composed of many distinct kinds of proteins, but most previous work analyzed the biofilm effect of loss of only a few. Here, we explored chemotaxis' role during biofilm formation in the human-associated pathogenic bacterium Helicobacter pylori. We found that chemotaxis proteins are involved in biofilm initiation in a manner that correlated with how they affected flagellar rotation. Biofilm initiation was high in mutants with counterclockwise (CCW) flagellar bias and low in those with clockwise bias. We supported the idea that a major driver of biofilm formation is flagellar rotational direction using a CCW-locked flagellar mutant, which stays CCW independent of chemotaxis input and showed elevated biofilm initiation. Our data suggest that CCW-rotating flagella, independent of chemotaxis inputs, are a biofilm-promoting signal.


Assuntos
Proteínas de Bactérias , Biofilmes , Quimiotaxia , Flagelos , Helicobacter pylori , Biofilmes/crescimento & desenvolvimento , Helicobacter pylori/fisiologia , Helicobacter pylori/genética , Flagelos/fisiologia , Flagelos/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Transdução de Sinais , Mutação , Rotação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA