Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Nature ; 609(7926): 335-340, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35853476

RESUMO

Adhesive pili assembled through the chaperone-usher pathway are hair-like appendages that mediate host tissue colonization and biofilm formation of Gram-negative bacteria1-3. Archaic chaperone-usher pathway pili, the most diverse and widespread chaperone-usher pathway adhesins, are promising vaccine and drug targets owing to their prevalence in the most troublesome multidrug-resistant pathogens1,4,5. However, their architecture and assembly-secretion process remain unknown. Here, we present the cryo-electron microscopy structure of the prototypical archaic Csu pilus that mediates biofilm formation of Acinetobacter baumannii-a notorious multidrug-resistant nosocomial pathogen. In contrast to the thick helical tubes of the classical type 1 and P pili, archaic pili assemble into an ultrathin zigzag architecture secured by an elegant clinch mechanism. The molecular clinch provides the pilus with high mechanical stability as well as superelasticity, a property observed for the first time, to our knowledge, in biomolecules, while enabling a more economical and faster pilus production. Furthermore, we demonstrate that clinch formation at the cell surface drives pilus secretion through the outer membrane. These findings suggest that clinch-formation inhibitors might represent a new strategy to fight multidrug-resistant bacterial infections.


Assuntos
Acinetobacter baumannii , Microscopia Crioeletrônica , Fímbrias Bacterianas , Chaperonas Moleculares , Acinetobacter baumannii/citologia , Acinetobacter baumannii/ultraestrutura , Elasticidade , Proteínas de Fímbrias/química , Proteínas de Fímbrias/metabolismo , Proteínas de Fímbrias/ultraestrutura , Fímbrias Bacterianas/química , Fímbrias Bacterianas/metabolismo , Fímbrias Bacterianas/ultraestrutura , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/ultraestrutura
2.
Mol Microbiol ; 117(3): 610-617, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34592048

RESUMO

Electron cryo-microscopy (cryo-EM) has lately emerged as a powerful method in structural biology and cell biology. While cryo-EM single-particle analysis (SPA) is now routinely delivering structures of purified proteins and protein complexes at near-atomic resolution, the use of electron cryo-tomography (cryo-ET), together with subtomogram averaging, is allowing visualization of macromolecular complexes in their native cellular environment, at unprecedented resolution. The unique ability of cryo-EM to provide information at many spatial resolution scales from ångströms to microns makes it an invaluable tool that bridges the classic "resolution-gap" between structural biology and cell biology domains. Like in many other fields of biology, in recent years, cryo-EM has revolutionized our understanding of pathogen biology, host-pathogen interaction and has made significant strides toward structure-based drug discovery. In a very recent example, during the ongoing coronavirus disease (COVID-19) pandemic, the structure of the stabilized severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein was deciphered by SPA. This led to the development of multiple vaccines. Alongside, cryo-ET provided key insights into the structure of the native virion, mechanism of its entry, replication, and budding; demonstrating the unrivaled power of cryo-EM in investigating pathogen biology, host-pathogen interaction, and drug discovery. In this review, we showcase a few examples of how different imaging modalities within cryo-EM have enabled the study of microbiology and host-pathogen interaction.


Assuntos
COVID-19 , SARS-CoV-2 , Biologia , Microscopia Crioeletrônica/métodos , Descoberta de Drogas , Interações Hospedeiro-Patógeno , Humanos
3.
EMBO J ; 38(14): e100957, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31304634

RESUMO

The self-assembly of cellular macromolecular machines such as the bacterial flagellar motor requires the spatio-temporal synchronization of gene expression with proper protein localization and association of dozens of protein components. In Salmonella and Escherichia coli, a sequential, outward assembly mechanism has been proposed for the flagellar motor starting from the inner membrane, with the addition of each new component stabilizing the previous one. However, very little is known about flagellar disassembly. Here, using electron cryo-tomography and sub-tomogram averaging of intact Legionella pneumophila, Pseudomonas aeruginosa, and Shewanella oneidensis cells, we study flagellar motor disassembly and assembly in situ. We first show that motor disassembly results in stable outer membrane-embedded sub-complexes. These sub-complexes consist of the periplasmic embellished P- and L-rings, and bend the membrane inward while it remains apparently sealed. Additionally, we also observe various intermediates of the assembly process including an inner-membrane sub-complex consisting of the C-ring, MS-ring, and export apparatus. Finally, we show that the L-ring is responsible for reshaping the outer membrane, a crucial step in the flagellar assembly process.


Assuntos
Bactérias/citologia , Proteínas de Bactérias/metabolismo , Flagelos/ultraestrutura , Bactérias/metabolismo , Bactérias/ultraestrutura , Membrana Externa Bacteriana/metabolismo , Tomografia com Microscopia Eletrônica , Escherichia coli/citologia , Escherichia coli/metabolismo , Escherichia coli/ultraestrutura , Flagelos/metabolismo , Legionella pneumophila/citologia , Legionella pneumophila/metabolismo , Legionella pneumophila/ultraestrutura , Pseudomonas aeruginosa/citologia , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/ultraestrutura , Shewanella/citologia , Shewanella/metabolismo , Shewanella/ultraestrutura
4.
J Bacteriol ; 204(8): e0014422, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35862756

RESUMO

The bacterial flagellar type III secretion system (fT3SS) is a suite of membrane-embedded and cytoplasmic proteins responsible for building the flagellar motility machinery. Homologous nonflagellar (NF-T3SS) proteins form the injectisome machinery that bacteria use to deliver effector proteins into eukaryotic cells, and other family members were recently reported to be involved in the formation of membrane nanotubes. Here, we describe a novel, evolutionarily widespread, hat-shaped structure embedded in the inner membranes of bacteria, of yet-unidentified function, that is present in species containing fT3SS. Mutant analysis suggests a relationship between this novel structure and the fT3SS, but not the NF-T3SS. While the function of this novel structure remains unknown, we hypothesize that either some of the fT3SS proteins assemble within the hat-like structure, perhaps including the fT3SS core complex, or that fT3SS components regulate other proteins that form part of this novel structure. IMPORTANCE The type III secretion system (T3SS) is a fascinating suite of proteins involved in building diverse macromolecular systems, including the bacterial flagellar motility machine, the injectisome machinery that bacteria use to inject effector proteins into host cells, and probably membrane nanotubes which connect bacterial cells. Here, we accidentally discovered a novel inner membrane-associated complex related to the flagellar T3SS. Examining our lab database, which is comprised of more than 40,000 cryo-tomograms of dozens of species, we discovered that this novel structure is both ubiquitous and ancient, being present in highly divergent classes of bacteria. Discovering a novel, widespread structure related to what are among the best-studied molecular machines in bacteria will open new venues for research aiming at understanding the function and evolution of T3SS proteins.


Assuntos
Flagelos , Sistemas de Secreção Tipo III , Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Estruturas Bacterianas , Flagelos/metabolismo , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo
5.
Biochem Soc Trans ; 50(1): 95-105, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35076655

RESUMO

Viruses can be enveloped or non-enveloped, and require a host cell to replicate and package their genomes into new virions to infect new cells. To accomplish this task, viruses hijack the host-cell machinery to facilitate their replication by subverting and manipulating normal host cell function. Enveloped viruses can have severe consequences for human health, causing various diseases such as acquired immunodeficiency syndrome (AIDS), seasonal influenza, COVID-19, and Ebola virus disease. The complex arrangement and pleomorphic architecture of many enveloped viruses pose a challenge for the more widely used structural biology techniques, such as X-ray crystallography. Cryo-electron tomography (cryo-ET), however, is a particularly well-suited tool for overcoming the limitations associated with visualizing the irregular shapes and morphology enveloped viruses possess at macromolecular resolution. The purpose of this review is to explore the latest structural insights that cryo-ET has revealed about enveloped viruses, with particular attention given to their architectures, mechanisms of entry, replication, assembly, maturation and egress during infection. Cryo-ET is unique in its ability to visualize cellular landscapes at 3-5 nanometer resolution. Therefore, it is the most suited technique to study asymmetric elements and structural rearrangements of enveloped viruses during infection in their native cellular context.


Assuntos
Vírus/ultraestrutura , Microscopia Crioeletrônica , Ebolavirus/metabolismo , Ebolavirus/ultraestrutura , Tomografia com Microscopia Eletrônica , HIV-1/metabolismo , HIV-1/ultraestrutura , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Humanos , SARS-CoV-2/metabolismo , SARS-CoV-2/ultraestrutura , Vírus/metabolismo
6.
J Bacteriol ; 203(3)2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33199282

RESUMO

Cellulose is a widespread component of bacterial biofilms, where its properties of exceptional water retention, high tensile strength, and stiffness prevent dehydration and mechanical disruption of the biofilm. Bacteria in the genus Gluconacetobacter secrete crystalline cellulose, with a structure very similar to that found in plant cell walls. How this higher-order structure is produced is poorly understood. We used cryo-electron tomography and focused-ion-beam milling of native bacterial biofilms to image cellulose-synthesizing Gluconacetobacter hansenii and Gluconacetobacter xylinus bacteria in a frozen-hydrated, near-native state. We confirm previous results suggesting that cellulose crystallization occurs serially following its secretion along one side of the cell, leading to a cellulose ribbon that can reach several micrometers in length and combine with ribbons from other cells to form a robust biofilm matrix. We were able to take direct measurements in a near-native state of the cellulose sheets. Our results also reveal a novel cytoskeletal structure, which we have named the cortical belt, adjacent to the inner membrane and underlying the sites where cellulose is seen emerging from the cell. We found that this structure is not present in other cellulose-synthesizing bacterial species, Agrobacterium tumefaciens and Escherichia coli 1094, which do not produce organized cellulose ribbons. We therefore propose that the cortical belt holds the cellulose synthase complexes in a line to form higher-order cellulose structures, such as sheets and ribbons.IMPORTANCE This work's relevance for the microbiology community is twofold. It delivers for the first time high-resolution near-native snapshots of Gluconacetobacter spp. (previously Komagataeibacter spp.) in the process of cellulose ribbon synthesis, in their native biofilm environment. It puts forward a noncharacterized cytoskeleton element associated with the side of the cell where the cellulose synthesis occurs. This represents a step forward in the understanding of the cell-guided process of crystalline cellulose synthesis, studied specifically in the Gluconacetobacter genus and still not fully understood. Additionally, our successful attempt to use cryo-focused-ion-beam milling through biofilms to image the cells in their native environment will drive the community to use this tool for the morphological characterization of other studied biofilms.


Assuntos
Celulose/ultraestrutura , Citoesqueleto/ultraestrutura , Gluconacetobacter/metabolismo , Gluconacetobacter/ultraestrutura , Acetobacteraceae/metabolismo , Acetobacteraceae/ultraestrutura , Biofilmes , Celulose/metabolismo , Cristalização , Citoesqueleto/metabolismo , Tomografia com Microscopia Eletrônica , Elétrons , Escherichia coli/metabolismo , Gluconacetobacter xylinus/metabolismo , Gluconacetobacter xylinus/ultraestrutura , Microfibrilas
7.
EMBO J ; 36(11): 1577-1589, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28438890

RESUMO

FtsZ, the bacterial homologue of eukaryotic tubulin, plays a central role in cell division in nearly all bacteria and many archaea. It forms filaments under the cytoplasmic membrane at the division site where, together with other proteins it recruits, it drives peptidoglycan synthesis and constricts the cell. Despite extensive study, the arrangement of FtsZ filaments and their role in division continue to be debated. Here, we apply electron cryotomography to image the native structure of intact dividing cells and show that constriction in a variety of Gram-negative bacterial cells, including Proteus mirabilis and Caulobacter crescentus, initiates asymmetrically, accompanied by asymmetric peptidoglycan incorporation and short FtsZ-like filament formation. These results show that a complete ring of FtsZ is not required for constriction and lead us to propose a model for FtsZ-driven division in which short dynamic FtsZ filaments can drive initial peptidoglycan synthesis and envelope constriction at the onset of cytokinesis, later increasing in length and number to encircle the division plane and complete constriction.


Assuntos
Proteínas de Bactérias/metabolismo , Caulobacter crescentus/citologia , Caulobacter crescentus/crescimento & desenvolvimento , Citocinese , Proteínas do Citoesqueleto/metabolismo , Multimerização Proteica , Proteus mirabilis/citologia , Proteus mirabilis/crescimento & desenvolvimento , Parede Celular/química , Parede Celular/metabolismo , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Peptidoglicano/análise , Peptidoglicano/biossíntese
8.
Proc Natl Acad Sci U S A ; 114(30): 8077-8082, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28696299

RESUMO

A recurrent emerging theme is the targeting of proteins to subcellular microdomains within bacterial cells, particularly to the poles. In most cases, it has been assumed that this localization is critical to the protein's function. Legionella pneumophila uses a type IVB secretion system (T4BSS) to export a large number of protein substrates into the cytoplasm of host cells. Here we show that the Legionella export apparatus is localized to the bacterial poles, as is consistent with many T4SS substrates being retained on the phagosomal membrane adjacent to the poles of the bacterium. More significantly, we were able to demonstrate that polar secretion of substrates is critically required for Legionella's alteration of the host endocytic pathway, an activity required for this pathogen's virulence.


Assuntos
Legionella pneumophila/fisiologia , Legionella pneumophila/patogenicidade , Sistemas de Secreção Tipo IV/metabolismo , Animais , Divisão Celular , Polaridade Celular , Feminino , Camundongos
9.
Traffic ; 18(2): 123-133, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27976831

RESUMO

The assembly of microtubule-based cytoskeleton propels the cilia and flagella growth. Previous studies have indicated that the kinesin-2 family motors transport tubulin into the cilia through intraflagellar transport. Here, we report a direct interaction between the C-terminal tail fragments of heterotrimeric kinesin-2 and α-tubulin1 isoforms in vitro. Blot overlay screen, affinity purification from tissue extracts, cosedimentation with subtilisin-treated microtubule and LC-ESI-MS/MS characterization of the tail-fragment-associated tubulin identified an association between the tail domains and α-tubulin1A/D isotype. The interaction was confirmed by Forster's resonance energy transfer assay in tissue-cultured cells. The overexpression of the recombinant tails in NIH3T3 cells affected the primary cilia growth, which was rescued by coexpression of a α-tubulin1 transgene. Furthermore, fluorescent recovery after photobleach analysis in the olfactory cilia of Drosophila indicated that tubulin is transported in a non-particulate form requiring kinesin-2. These results provide additional new insight into the mechanisms underlying selective tubulin isoform enrichment in the cilia.


Assuntos
Cílios/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Subunidades Proteicas/metabolismo , Tubulina (Proteína)/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico/fisiologia , Drosophila , Flagelos/metabolismo , Cinesinas , Camundongos , Microtúbulos/metabolismo , Células NIH 3T3 , Ligação Proteica/fisiologia , Domínios Proteicos/fisiologia
10.
EMBO J ; 34(18): 2312-20, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26271102

RESUMO

It is now well established that prokaryotic cells assemble diverse proteins into dynamic cytoskeletal filaments that perform essential cellular functions. Although most of the filaments assemble on their own to form higher order structures, growing evidence suggests that there are a number of prokaryotic proteins that polymerise only in the presence of a matrix such as DNA, lipid membrane or even another filament. Matrix-assisted filament systems are frequently nucleotide dependent and cytomotive but rarely considered as part of the bacterial cytoskeleton. Here, we categorise this family of filament-forming systems as collaborative filaments and introduce a simple nomenclature. Collaborative filaments are frequent in both eukaryotes and prokaryotes and are involved in vital cellular processes including chromosome segregation, DNA repair and maintenance, gene silencing and cytokinesis to mention a few. In this review, we highlight common principles underlying collaborative filaments and correlate these with known functions.


Assuntos
Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Bactérias/genética , Proteínas de Bactérias/genética , Divisão Celular/fisiologia , Proteínas do Citoesqueleto/genética , Citoesqueleto/genética , Reparo do DNA/fisiologia , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Inativação Gênica/fisiologia , Humanos
11.
EMBO Rep ; 18(5): 726-732, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28336774

RESUMO

Type IV secretion systems (T4SSs) are large macromolecular machines that translocate protein and DNA and are involved in the pathogenesis of multiple human diseases. Here, using electron cryotomography (ECT), we report the in situ structure of the Dot/Icm type IVB secretion system (T4BSS) utilized by the human pathogen Legionella pneumophila This is the first structure of a type IVB secretion system, and also the first structure of any T4SS in situ While the Dot/Icm system shares almost no sequence similarity with type IVA secretion systems (T4ASSs), its overall structure is seen here to be remarkably similar to previously reported T4ASS structures (those encoded by the R388 plasmid in Escherichia coli and the cag pathogenicity island in Helicobacter pylori). This structural similarity suggests shared aspects of mechanism. However, compared to the negative-stain reconstruction of the purified T4ASS from the R388 plasmid, the L. pneumophila Dot/Icm system is approximately twice as long and wide and exhibits several additional large densities, reflecting type-specific elaborations and potentially better structural preservation in situ.


Assuntos
Tomografia com Microscopia Eletrônica/métodos , Legionella pneumophila/química , Sistemas de Secreção Tipo IV/química , Sistemas de Secreção Tipo IV/ultraestrutura , Proteínas de Bactérias/química , Proteínas de Bactérias/ultraestrutura , Regulação Bacteriana da Expressão Gênica , Legionella pneumophila/genética , Legionella pneumophila/patogenicidade , Legionella pneumophila/ultraestrutura , Plasmídeos
12.
EMBO Rep ; 18(9): 1660-1670, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28729461

RESUMO

Archaeal swimming motility is driven by archaella: rotary motors attached to long extracellular filaments. The structure of these motors, and particularly how they are anchored in the absence of a peptidoglycan cell wall, is unknown. Here, we use electron cryotomography to visualize the archaellar basal body in vivo in Thermococcus kodakaraensis KOD1. Compared to the homologous bacterial type IV pilus (T4P), we observe structural similarities as well as several unique features. While the position of the cytoplasmic ATPase appears conserved, it is not braced by linkages that extend upward through the cell envelope as in the T4P, but rather by cytoplasmic components that attach it to a large conical frustum up to 500 nm in diameter at its base. In addition to anchoring the lophotrichous bundle of archaella, the conical frustum associates with chemosensory arrays and ribosome-excluding material and may function as a polar organizing center for the coccoid cells.


Assuntos
Extensões da Superfície Celular/ultraestrutura , Citoplasma/ultraestrutura , Thermococcus/fisiologia , Thermococcus/ultraestrutura , Adenosina Trifosfatases/metabolismo , Proteínas Arqueais/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Extensões da Superfície Celular/fisiologia , Microscopia Crioeletrônica , Citoplasma/fisiologia , Flagelos/fisiologia , Flagelos/ultraestrutura , Thermococcus/citologia
13.
Subcell Biochem ; 84: 213-244, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28500527

RESUMO

In bacteria and archaea, the most widespread cell division system is based on the tubulin homologue FtsZ protein, whose filaments form the cytokinetic Z-ring. FtsZ filaments are tethered to the membrane by anchors such as FtsA and SepF and are regulated by accessory proteins. One such set of proteins is responsible for Z-ring's spatiotemporal regulation, essential for the production of two equal-sized daughter cells. Here, we describe how our still partial understanding of the FtsZ-based cell division process has been progressed by visualising near-atomic structures of Z-rings and complexes that control Z-ring positioning in cells, most notably the MinCDE and Noc systems that act by negatively regulating FtsZ filaments. We summarise available data and how they inform mechanistic models for the cell division process.


Assuntos
Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/metabolismo , Divisão Celular , Citoesqueleto
14.
Traffic ; 13(7): 979-91, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22486887

RESUMO

Bulk flow constitutes a substantial part of the slow transport of soluble proteins in axons. Though the underlying mechanism is unclear, evidences indicate that intermittent, kinesin-based movement of large protein-aggregates aids this process. Choline acetyltransferase (ChAT), a soluble enzyme catalyzing acetylcholine synthesis, propagates toward the synapse at an intermediate, slow rate. The presynaptic enrichment of ChAT requires heterotrimeric kinesin-2, comprising KLP64D, KLP68D and DmKAP, in Drosophila. Here, we show that the bulk flow of a recombinant Green Fluorescent Protein-tagged ChAT (GFP::ChAT), in Drosophila axons, lacks particulate features. It occurs for a brief period during the larval stages. In addition, both the endogenous ChAT and GFP::ChAT directly bind to the KLP64D tail, which is essential for the GFP::ChAT entry and anterograde flow in axon. These evidences suggest that a direct interaction with motor proteins could regulate the bulk flow of soluble proteins, and thus establish their asymmetric distribution.


Assuntos
Transporte Axonal/fisiologia , Colina O-Acetiltransferase/metabolismo , Proteínas de Drosophila/metabolismo , Cinesinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Axonal/genética , Proteínas de Transporte/metabolismo , Neurônios Colinérgicos/enzimologia , Neurônios Colinérgicos/metabolismo , Drosophila/enzimologia , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/química , Recuperação de Fluorescência Após Fotodegradação , Cinesinas/química , Larva/enzimologia , Larva/metabolismo , Proteínas Associadas aos Microtúbulos/química , Domínios e Motivos de Interação entre Proteínas , Sinapses/enzimologia , Sinapses/metabolismo
15.
J Phys Chem Lett ; 15(4): 919-924, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38241259

RESUMO

The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the SARS-CoV-2 coronavirus and the perpetual rise of new variants warrant investigation of the molecular and structural details of the infection process and modulation of the host defense by viral proteins. This Letter reports the combined experimental and computational approaches to provide key insights into the structural and functional basis of Nsp1's association with different cyclophilins and FKBPs in regulating COVID-19 infection. We demonstrated the real-time stability and functional dynamics of the Nsp1-CypA/FKBP1A complex and investigated the repurposing of potential inhibitors that could block these interactions. Overall, we provided insights into the inhibitory role Nsp1 in downstream interferon production, a key aspect for host defense that prevents the SARS-CoV-2 or related family of corona virus infection.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Ciclofilinas , Proteínas não Estruturais Virais/metabolismo , Interferons
16.
Microbiologyopen ; 13(2): e1401, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38409911

RESUMO

Prevotella intermedia, a Gram-negative bacterium from the Bacteroidota phylum, is associated with periodontitis. Other species within this phylum are known to possess the general O-glycosylation system. The O-glycoproteome has been characterized in several species, including Tannerella forsythia, Porphyromonas gingivalis, and Flavobacterium johnsoniae. In our study, we used electron cryotomography (cryoET) and glycoproteomics to reveal the ultrastructure of P. intermedia and characterize its O-glycoproteome. Our cryoET analysis unveiled the ultrastructural details of the cell envelope and outer membrane vesicles (OMVs) of P. intermedia. We observed an electron-dense surface layer surrounding both cells and OMVs. The OMVs were often large (>200 nm) and presented two types, with lumens being either electron-dense or translucent. LC-MS/MS analyses of P. intermedia fractions led to the identification of 1655 proteins, which included 62 predicted T9SS cargo proteins. Within the glycoproteome, we identified 443 unique O-glycosylation sites within 224 glycoproteins. Interestingly, the O-glycosylation motif exhibited a broader range than reported in other species, with O-glycosylation found at D(S/T)(A/I/L/M/T/V/S/C/G/F/N/E/Q/D/P). We identified a single O-glycan with a delta mass of 1531.48 Da. Its sequence was determined by MS2 and MS3 analyses using both collision-induced dissociation and high-energy collisional dissociation fragmentation modes. After partial deglycosylation with trifluoromethanesulfonic acid, the O-glycan sequence was confirmed to be dHex-dHex-HexNAc (HPO3 -C6 H12 O5 )-dHex-Hex-HexA-Hex(dHex). Bioinformatic analyses predicted the localization of O-glycoproteins, with 73 periplasmic proteins, 53 inner membrane proteins, 52 lipoproteins, 26 outer membrane proteins, and 14 proteins secreted by the T9SS.


Assuntos
Glicoproteínas , Espectrometria de Massas em Tandem , Glicosilação , Prevotella intermedia/metabolismo , Cromatografia Líquida , Espectrometria de Massas em Tandem/métodos , Proteínas de Membrana/metabolismo , Proteoma/metabolismo , Polissacarídeos
17.
Environ Microbiol Rep ; 16(2): e13258, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38589217

RESUMO

DNA methylation serves a variety of functions across all life domains. In this study, we investigated archaeal methylomics within a tripartite xylanolytic halophilic consortium. This consortium includes Haloferax lucertense SVX82, Halorhabdus sp. SVX81, and an ectosymbiotic Candidatus Nanohalococcus occultus SVXNc, a nano-sized archaeon from the DPANN superphylum. We utilized PacBio SMRT and Illumina cDNA sequencing to analyse samples from consortia of different compositions for methylomics and transcriptomics. Endogenous cTAG methylation, typical of Haloferax, was accompanied in this strain by methylation at four other motifs, including GDGcHC methylation, which is specific to the ectosymbiont. Our analysis of the distribution of methylated and unmethylated motifs suggests that autochthonous cTAG methylation may influence gene regulation. The frequency of GRAGAaG methylation increased in highly expressed genes, while CcTTG and GTCGaGG methylation could be linked to restriction-modification (RM) activity. Generally, the RM activity might have been reduced during the evolution of this archaeon to balance the protection of cells from intruders, the reduction of DNA damage due to self-restriction in stressful environments, and the benefits of DNA exchange under extreme conditions. Our methylomics, transcriptomics and complementary electron cryotomography (cryo-ET) data suggest that the nanohaloarchaeon exports its methyltransferase to methylate the Haloferax genome, unveiling a new aspect of the interaction between the symbiont and its host.


Assuntos
Archaea , Metilação de DNA , Archaea/genética , Perfilação da Expressão Gênica , Expressão Gênica , Metiltransferases/genética , DNA Arqueal/genética
18.
Commun Biol ; 7(1): 461, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38627519

RESUMO

EphB6 is an understudied ephrin receptor tyrosine pseudokinase that is downregulated in multiple types of metastatic cancers. Unlike its kinase-active counterparts which autophosphorylate and transmit signals upon intercellular interaction, little is known about how EphB6 functions in the absence of intrinsic kinase activity. Here, we unveil a molecular mechanism of cell-cell interaction driven by EphB6. We identify ephrinB1 as a cognate ligand of EphB6 and show that in trans interaction of EphB6 with ephrinB1 on neighboring cells leads to the formation of large co-clusters at the plasma membrane. These co-clusters exhibit a decreased propensity towards endocytosis, suggesting a unique characteristic for this type of cell-cell interaction. Using lattice light-sheet microscopy, 3D structured illumination microscopy and cryo-electron tomography techniques, we show that co-clustering of EphB6 and ephrinB1 promotes the formation of double-membrane tubular structures between cells. Importantly, we also demonstrate that these intercellular structures stabilize cell-cell adhesion, leading to a reduction in the invasive behavior of cancer cells. Our findings rationalize a role for EphB6 pseudokinase as a tumor suppressor when interacting with its ligands in trans.


Assuntos
Fosforilação , Invasividade Neoplásica
19.
ISME J ; 18(1)2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38470311

RESUMO

Many bacteria produce antimicrobial compounds such as lantibiotics to gain advantage in the competitive natural environments of microbiomes. Epilancins constitute an until now underexplored family of lantibiotics with an unknown ecological role and unresolved mode of action. We discovered production of an epilancin in the nasal isolate Staphylococcus epidermidis A37. Using bioinformatic tools, we found that epilancins are frequently encoded within staphylococcal genomes, highlighting their ecological relevance. We demonstrate that production of epilancin A37 contributes to Staphylococcus epidermidis competition specifically against natural corynebacterial competitors. Combining microbiological approaches with quantitative in vivo and in vitro fluorescence microscopy and cryo-electron tomography, we show that A37 enters the corynebacterial cytoplasm through a partially transmembrane-potential-driven uptake without impairing the cell membrane function. Upon intracellular aggregation, A37 induces the formation of intracellular membrane vesicles, which are heavily loaded with the compound and are essential for the antibacterial activity of the epilancin. Our work sheds light on the ecological role of epilancins for staphylococci mediated by a mode of action previously unknown for lantibiotics.


Assuntos
Bacteriocinas , Microbiota , Bacteriocinas/farmacologia , Staphylococcus epidermidis/metabolismo , Staphylococcus , Antibacterianos/farmacologia , Antibacterianos/metabolismo
20.
ISME J ; 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39113594

RESUMO

DPANN archaea are an enigmatic superphylum that are difficult to isolate and culture in the laboratory due to their specific culture conditions and apparent ectosymbiotic lifestyle. Here we successfully isolated and cultivated a co-culture system of a novel Nanobdellota archaeon YN1 and its host Sulfurisphaera ohwakuensis YN1HA. We characterised the co-culture system by complementary methods, including metagenomics and metabolic pathway analysis, fluorescence microscopy, and high-resolution electron cryo-tomography (cryoET). We show that YN1 is deficient in essential metabolic processes and requires host resources to proliferate. CryoET imaging revealed an enormous attachment organelle present in the YN1 envelope that forms a direct interaction with the host cytoplasm, bridging the two cells. Together our results unravel the molecular and structural basis of ectosymbiotic relationship between YN1 and YNHA. This research broadens our understanding of DPANN biology and the versatile nature of their ectosymbiotic relationships.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA