Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Immunity ; 49(1): 120-133.e9, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30005826

RESUMO

B lymphocytes can suppress immunity through interleukin (IL)-10 production in infectious, autoimmune, and malignant diseases. Here, we have identified a natural plasma cell subset that distinctively expresses the inhibitory receptor LAG-3 and mediates this function in vivo. These plasma cells also express the inhibitory receptors CD200, PD-L1, and PD-L2. They develop from various B cell subsets in a B cell receptor (BCR)-dependent manner independently of microbiota in naive mice. After challenge they upregulate IL-10 expression via a Toll-like receptor-driven mechanism within hours and without proliferating. This function is associated with a unique transcriptome and epigenome, including the lowest amount of DNA methylation at the Il10 locus compared to other B cell subsets. Their augmented accumulation in naive mutant mice with increased BCR signaling correlates with the inhibition of memory T cell formation and vaccine efficacy after challenge. These natural regulatory plasma cells may be of broad relevance for disease intervention.


Assuntos
Antígenos CD/genética , Expressão Gênica , Interleucina-10/biossíntese , Plasmócitos/imunologia , Animais , Antígenos CD/imunologia , Subpopulações de Linfócitos B/imunologia , Epigênese Genética , Feminino , Perfilação da Expressão Gênica , Interleucina-10/genética , Ativação Linfocitária , Masculino , Camundongos , Plasmócitos/fisiologia , Receptores de Antígenos de Linfócitos B/metabolismo , Salmonelose Animal/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Receptores Toll-Like/metabolismo , Regulação para Cima/genética , Vacinas/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
2.
Development ; 149(18)2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36093870

RESUMO

Proven roles for hemocytes (blood cells) have expanded beyond the control of infections in Drosophila. Despite this, the crucial role of hemocytes in post-embryonic development has long thought to be limited to control of microorganisms during metamorphosis. This has previously been shown by rescue of adult development in hemocyte-ablation models under germ-free conditions. Here, we show that hemocytes have an essential role in post-embryonic development beyond their ability to control the microbiota. Using a newly generated strong hemocyte-specific driver line for the GAL4/UAS system, we show that specific ablation of hemocytes is early pupal lethal, even under axenic conditions. Genetic rescue experiments prove that this is a hemocyte-specific phenomenon. RNA-seq data suggests that dysregulation of the midgut is a prominent consequence of hemocyte ablation in larval stages, resulting in reduced gut lengths. Dissection suggests that multiple processes may be affected during metamorphosis. We believe this previously unreported role for hemocytes during metamorphosis is a major finding for the field.


Assuntos
Proteínas de Drosophila , Microbiota , Animais , Drosophila , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Desenvolvimento Embrionário , Hemócitos , Larva
3.
Gastroenterology ; 161(2): 623-636.e16, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33957136

RESUMO

BACKGROUND & AIMS: The homeostasis of the gastrointestinal epithelium relies on cell regeneration and differentiation into distinct lineages organized inside glands and crypts. Regeneration depends on Wnt/ß-catenin pathway activation, but to understand homeostasis and its dysregulation in disease, we need to identify the signaling microenvironment governing cell differentiation. By using gastric glands as a model, we have identified the signals inducing differentiation of surface mucus-, zymogen-, and gastric acid-producing cells. METHODS: We generated mucosoid cultures from the human stomach and exposed them to different growth factors to obtain cells with features of differentiated foveolar, chief, and parietal cells. We localized the source of the growth factors in the tissue of origin. RESULTS: We show that epidermal growth factor is the major fate determinant distinguishing the surface and inner part of human gastric glands. In combination with bone morphogenetic factor/Noggin signals, epidermal growth factor controls the differentiation of foveolar cells vs parietal or chief cells. We also show that epidermal growth factor is likely to underlie alteration of the gastric mucosa in the precancerous condition atrophic gastritis. CONCLUSIONS: Use of our recently established mucosoid cultures in combination with analysis of the tissue of origin provided a robust strategy to understand differentiation and patterning of human tissue and allowed us to draw a new, detailed map of the signaling microenvironment in the human gastric glands.


Assuntos
Padronização Corporal/efeitos dos fármacos , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/efeitos dos fármacos , Mucosa Gástrica/efeitos dos fármacos , Proteínas de Transporte/farmacologia , Linhagem da Célula , Células Cultivadas , Microambiente Celular , Celulas Principais Gástricas/efeitos dos fármacos , Celulas Principais Gástricas/metabolismo , Celulas Principais Gástricas/ultraestrutura , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Mucosa Gástrica/metabolismo , Mucosa Gástrica/ultraestrutura , Gastrite Atrófica/metabolismo , Gastrite Atrófica/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Organoides , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/ultraestrutura , Via de Sinalização Wnt
4.
Proc Natl Acad Sci U S A ; 115(32): E7568-E7577, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30038005

RESUMO

Mosquito blood cells are immune cells that help control infection by vector-borne pathogens. Despite their importance, little is known about mosquito blood cell biology beyond morphological and functional criteria used for their classification. Here, we combined the power of single-cell RNA sequencing, high-content imaging flow cytometry, and single-molecule RNA hybridization to analyze a subset of blood cells of the malaria mosquito Anopheles gambiae By demonstrating that blood cells express nearly half of the mosquito transcriptome, our dataset represents an unprecedented view into their transcriptional program. Analyses of differentially expressed genes identified transcriptional signatures of two cell types and provide insights into the current classification of these cells. We further demonstrate the active transfer of a cellular marker between blood cells that may confound their identification. We propose that cell-to-cell exchange may contribute to cellular diversity and functional plasticity seen across biological systems.


Assuntos
Anopheles/genética , Células Sanguíneas/classificação , Plasticidade Celular/genética , Malária/transmissão , Mosquitos Vetores/genética , Animais , Animais Geneticamente Modificados , Anopheles/imunologia , Células Sanguíneas/imunologia , Comunicação Celular/genética , Conjuntos de Dados como Assunto , Feminino , Genômica/métodos , Mosquitos Vetores/imunologia , RNA/genética , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Transcriptoma
5.
Nature ; 512(7515): 387-92, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25119038

RESUMO

The aryl hydrocarbon receptor (AhR) is a highly conserved ligand-dependent transcription factor that senses environmental toxins and endogenous ligands, thereby inducing detoxifying enzymes and modulating immune cell differentiation and responses. We hypothesized that AhR evolved to sense not only environmental pollutants but also microbial insults. We characterized bacterial pigmented virulence factors, namely the phenazines from Pseudomonas aeruginosa and the naphthoquinone phthiocol from Mycobacterium tuberculosis, as ligands of AhR. Upon ligand binding, AhR activation leads to virulence factor degradation and regulated cytokine and chemokine production. The relevance of AhR to host defence is underlined by heightened susceptibility of AhR-deficient mice to both P. aeruginosa and M. tuberculosis. Thus, we demonstrate that AhR senses distinct bacterial virulence factors and controls antibacterial responses, supporting a previously unidentified role for AhR as an intracellular pattern recognition receptor, and identify bacterial pigments as a new class of pathogen-associated molecular patterns.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Mycobacterium tuberculosis/imunologia , Pigmentos Biológicos/metabolismo , Pseudomonas aeruginosa/imunologia , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Animais , Antibacterianos/metabolismo , Células da Medula Óssea/citologia , Citocinas/imunologia , Citocinas/metabolismo , Retroalimentação Fisiológica , Humanos , Ligantes , Ativação de Macrófagos , Camundongos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/metabolismo , Fenazinas/metabolismo , Pigmentos Biológicos/química , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Piocianina/metabolismo , Fatores de Virulência/química , Fatores de Virulência/metabolismo
6.
Gut ; 68(3): 400-413, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-29467166

RESUMO

OBJECTIVE: Helicobacter pylori causes life-long colonisation of the gastric mucosa, leading to chronic inflammation with increased risk of gastric cancer. Research on the pathogenesis of this infection would strongly benefit from an authentic human in vitro model. DESIGN: Antrum-derived gastric glands from surgery specimens served to establish polarised epithelial monolayers via a transient air-liquid interface culture stage to study cross-talk with H. pylori and the adjacent stroma. RESULTS: The resulting 'mucosoid cultures', so named because they recapitulate key characteristics of the gastric mucosa, represent normal stem cell-driven cultures that can be passaged for months. These highly polarised columnar epithelial layers encompass the various gastric antral cell types and secrete mucus at the apical surface. By default, they differentiate towards a foveolar, MUC5AC-producing phenotype, whereas Wnt signalling stimulates proliferation of MUC6-producing cells and preserves stemness-reminiscent of the gland base. Stromal cells from the lamina propria secrete Wnt inhibitors, antagonising stem-cell niche signalling and inducing differentiation. On infection with H. pylori, a strong inflammatory response is induced preferentially in the undifferentiated basal cell phenotype. Infection of cultures for several weeks produces foci of viable bacteria and a persistent inflammatory condition, while the secreted mucus establishes a barrier that only few bacteria manage to overcome. CONCLUSION: Gastric mucosoid cultures faithfully reproduce the features of normal human gastric epithelium, enabling new approaches for investigating the interaction of H. pylori with the epithelial surface and the cross-talk with the basolateral stromal compartment. Our observations provide striking insights in the regulatory circuits of inflammation and defence.


Assuntos
Mucosa Gástrica/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Infecções por Helicobacter/metabolismo , Homeostase/fisiologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Muco/metabolismo , Antro Pilórico/metabolismo , Antro Pilórico/microbiologia , Antro Pilórico/patologia , Nicho de Células-Tronco , Células Estromais/fisiologia , Técnicas de Cultura de Tecidos/métodos
7.
Nature ; 486(7402): 276-9, 2012 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-22699623

RESUMO

Pathogenic bacteria using a type III secretion system (T3SS) to manipulate host cells cause many different infections including Shigella dysentery, typhoid fever, enterohaemorrhagic colitis and bubonic plague. An essential part of the T3SS is a hollow needle-like protein filament through which effector proteins are injected into eukaryotic host cells. Currently, the three-dimensional structure of the needle is unknown because it is not amenable to X-ray crystallography and solution NMR, as a result of its inherent non-crystallinity and insolubility. Cryo-electron microscopy combined with crystal or solution NMR subunit structures has recently provided a powerful hybrid approach for studying supramolecular assemblies, resulting in low-resolution and medium-resolution models. However, such approaches cannot deliver atomic details, especially of the crucial subunit-subunit interfaces, because of the limited cryo-electron microscopic resolution obtained in these studies. Here we report an alternative approach combining recombinant wild-type needle production, solid-state NMR, electron microscopy and Rosetta modelling to reveal the supramolecular interfaces and ultimately the complete atomic structure of the Salmonella typhimurium T3SS needle. We show that the 80-residue subunits form a right-handed helical assembly with roughly 11 subunits per two turns, similar to that of the flagellar filament of S. typhimurium. In contrast to established models of the needle in which the amino terminus of the protein subunit was assumed to be α-helical and positioned inside the needle, our model reveals an extended amino-terminal domain that is positioned on the surface of the needle, while the highly conserved carboxy terminus points towards the lumen.


Assuntos
Sistemas de Secreção Bacterianos , Modelos Moleculares , Salmonella typhimurium/química , Células HeLa , Humanos , Microscopia Eletrônica , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína
8.
Chembiochem ; 18(10): 888-893, 2017 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-28240414

RESUMO

Neutrophils are short-lived leukocytes that migrate to sites of infection as part of the acute immune response, where they phagocytose, degranulate, and form neutrophil extracellular traps (NETs). During NET formation, the nuclear lobules of neutrophils disappear and the chromatin expands and, accessorized with neutrophilic granule proteins, is expelled. NETs can be pathogenic in, for example, sepsis, cancer, and autoimmune and cardiovascular diseases. Therefore, the identification of inhibitors of NET formation is of great interest. Screening of a focused library of natural-product-inspired compounds by using a previously validated phenotypic NET assay identified a group of tetrahydroisoquinolines as new NET formation inhibitors. This compound class opens up new avenues for the study of cellular death through NET formation (NETosis) at different stages, and might inspire new medicinal chemistry programs aimed at NET-dependent diseases.


Assuntos
Armadilhas Extracelulares/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Neutrófilos/metabolismo , Tetra-Hidroisoquinolinas/farmacologia , Morte Celular , Armadilhas Extracelulares/efeitos dos fármacos , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/metabolismo , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos
9.
Traffic ; 15(4): 362-82, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24423236

RESUMO

For membrane-bound intracellular pathogens, the surrounding vacuole is the portal of communication with the host cell. The parasitophorous vacuole (PV) harboring intrahepatocytic Plasmodium parasites satisfies the parasites' needs of nutrition and protection from host defenses to allow the rapid parasite growth that occurs during the liver stage of infection. In this study, we visualized the PV membrane (PVM) and the associated tubovesicular network (TVN) through fluorescent tagging of two PVM-resident Plasmodium berghei proteins, UIS4 and IBIS1. This strategy revealed previously unrecognized dynamics with which these membranes extend throughout the host cell. We observed dynamic vesicles, elongated clusters of membranes and long tubules that rapidly extend and contract from the PVM in a microtubule-dependent manner. Live microscopy, correlative light-electron microscopy and fluorescent recovery after photobleaching enabled a detailed characterization of these membranous features, including velocities, the distribution of UIS4 and IBIS1, and the connectivity of PVM and TVN. Labeling of host cell compartments revealed association of late endosomes and lysosomes with the elongated membrane clusters. Moreover, the signature host autophagosome protein LC3 was recruited to the PVM and TVN and colocalized with UIS4. Together, our data demonstrate that the membranes surrounding intrahepatic Plasmodium are involved in active remodeling of host cells.


Assuntos
Fígado/parasitologia , Plasmodium/metabolismo , Animais , Membrana Celular/metabolismo , Interações Hospedeiro-Parasita , Plasmodium/patogenicidade
10.
PLoS Pathog ; 10(1): e1003881, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24453973

RESUMO

The Type III Secretion System (T3SS) is a macromolecular complex used by Gram-negative bacteria to secrete effector proteins from the cytoplasm across the bacterial envelope in a single step. For many pathogens, the T3SS is an essential virulence factor that enables the bacteria to interact with and manipulate their respective host. A characteristic structural feature of the T3SS is the needle complex (NC). The NC resembles a syringe with a basal body spanning both bacterial membranes and a long needle-like structure that protrudes from the bacterium. Based on the paradigm of a syringe-like mechanism, it is generally assumed that effectors and translocators are unfolded and secreted from the bacterial cytoplasm through the basal body and needle channel. Despite extensive research on T3SS, this hypothesis lacks experimental evidence and the mechanism of secretion is not fully understood. In order to elucidate details of the T3SS secretion mechanism, we generated fusion proteins consisting of a T3SS substrate and a bulky protein containing a knotted motif. Because the knot cannot be unfolded, these fusions are accepted as T3SS substrates but remain inside the NC channel and obstruct the T3SS. To our knowledge, this is the first time substrate fusions have been visualized together with isolated NCs and we demonstrate that substrate proteins are secreted directly through the channel with their N-terminus first. The channel physically encloses the fusion protein and shields it from a protease and chemical modifications. Our results corroborate an elementary understanding of how the T3SS works and provide a powerful tool for in situ-structural investigations in the future. This approach might also be applicable to other protein secretion systems that require unfolding of their substrates prior to secretion.


Assuntos
Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/fisiologia , Citoplasma/metabolismo , Shigella flexneri/metabolismo , Proteínas de Bactérias/genética , Citoplasma/genética , Transporte Proteico/fisiologia , Shigella flexneri/genética , Shigella flexneri/ultraestrutura
11.
bioRxiv ; 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38558991

RESUMO

The bacterial flagellum is an organelle utilized by many Gram-negative bacteria to facilitate motility. The flagellum is composed of a several µm long, extracellular filament that is connected to a cytoplasmic rotor-stator complex via a periplasmic rod. Composed of ∼20 structural proteins, ranging from a few subunits to several thousand building blocks, the flagellum is a paradigm of a complex macromolecular structure that utilizes a highly regulated assembly process. This process is governed by multiple checkpoints that ensure an ordered gene expression pattern coupled to the assembly of the various flagellar building blocks in order to produce a functional flagellum. Using epifluorescence, super-resolution STED and transmission electron microscopy, we discovered that in Salmonella , the absence of one periplasmic protein, FlhE, prevents proper flagellar morphogenesis and results in the formation of periplasmic flagella. The periplasmic flagella disrupt cell wall synthesis, leading to a loss of the standard cell morphology resulting in cell lysis. We propose a model where FlhE functions as a periplasmic chaperone to control assembly of the periplasmic rod to prevent formation of periplasmic flagella. Our results highlight that bacteria evolved sophisticated regulatory mechanisms to control proper flagellar assembly and minor deviations from this highly regulated process can cause dramatic physiological consequences.

12.
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
13.
bioRxiv ; 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38979141

RESUMO

The flagellum is the most complex macromolecular structure known in bacteria and comprised of around two dozen distinct proteins. The main building block of the long, external flagellar filament, flagellin, is secreted through the flagellar type-III secretion system at a remarkable rate of several tens of thousands amino acids per second, significantly surpassing the rates achieved by other pore-based protein secretion systems. The evolutionary implications and potential benefits of this high secretion rate for flagellum assembly and function, however, have remained elusive. In this study, we provide both experimental and theoretical evidence that the flagellar secretion rate has been evolutionarily optimized to facilitate rapid and efficient construction of a functional flagellum. By synchronizing flagellar assembly, we found that a minimal filament length of 2.5 µm was required for swimming motility. Biophysical modelling revealed that this minimal filament length threshold resulted from an elasto-hydrodynamic instability of the whole swimming cell, dependent on the filament length. Furthermore, we developed a stepwise filament labeling method combined with electron microscopy visualization to validate predicted flagellin secretion rates of up to 10,000 amino acids per second. A biophysical model of flagellum growth demonstrates that the observed high flagellin secretion rate efficiently balances filament elongation and energy consumption, thereby enabling motility in the shortest amount of time. Taken together, these insights underscore the evolutionary pressures that have shaped the development and optimization of the flagellum and type-III secretion system, illuminating the intricate interplay between functionality and efficiency in assembly of large macromolecular structures. Significance statement: Our study demonstrates how protein secretion of the bacterial flagellum is finely tuned to optimize filament assembly rate and flagellum function while minimizing energy consumption. By measuring flagellar filament lengths and bacterial swimming after initiation of flag-ellum assembly, we were able to establish the minimal filament length necessary for swimming motility, which we rationalized physically as resulting from an elasto-hydrodynamic instability of the swimming cell. Our bio-physical model of flagellum growth further illustrates how the physiological flagellin secretion rate is optimized to maximize filament elongation while conserving energy. These findings illuminate the evolutionary pressures that have shaped the function of the bacterial flagellum and type-III secretion system, driving improvements in bacterial motility and overall fitness.

14.
J Biol Chem ; 287(4): 2410-22, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22139844

RESUMO

Plasmodium sporozoites, single cell eukaryotic pathogens, use their own actin/myosin-based motor machinery for life cycle progression, which includes forward locomotion, penetration of cellular barriers, and invasion of target cells. To display fast gliding motility, the parasite uses a high turnover of actin polymerization and adhesion sites. Paradoxically, only a few classic actin regulatory proteins appear to be encoded in the Plasmodium genome. Small heat shock proteins have been associated with cytoskeleton modulation in various biological processes. In this study, we identify HSP20 as a novel player in Plasmodium motility and provide molecular genetics evidence for a critical role of a small heat shock protein in cell traction and motility. We demonstrate that HSP20 ablation profoundly affects sporozoite-substrate adhesion, which translates into aberrant speed and directionality in vitro. Loss of HSP20 function impairs migration in the host, an important sporozoite trait required to find a blood vessel and reach the liver after being deposited in the skin by the mosquito. Our study also shows that fast locomotion of sporozoites is crucial during natural malaria transmission.


Assuntos
Proteínas de Choque Térmico HSP20/metabolismo , Locomoção/fisiologia , Plasmodium berghei/metabolismo , Proteínas de Protozoários/metabolismo , Esporozoítos/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Proteínas de Choque Térmico HSP20/genética , Malária/genética , Malária/metabolismo , Malária/transmissão , Camundongos , Plasmodium berghei/genética , Proteínas de Protozoários/genética
15.
J Cell Biol ; 176(2): 231-41, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17210947

RESUMO

Neutrophil extracellular traps (NETs) are extracellular structures composed of chromatin and granule proteins that bind and kill microorganisms. We show that upon stimulation, the nuclei of neutrophils lose their shape, and the eu- and heterochromatin homogenize. Later, the nuclear envelope and the granule membranes disintegrate, allowing the mixing of NET components. Finally, the NETs are released as the cell membrane breaks. This cell death process is distinct from apoptosis and necrosis and depends on the generation of reactive oxygen species (ROS) by NADPH oxidase. Patients with chronic granulomatous disease carry mutations in NADPH oxidase and cannot activate this cell-death pathway or make NETs. This novel ROS-dependent death allows neutrophils to fulfill their antimicrobial function, even beyond their lifespan.


Assuntos
Apoptose/fisiologia , Imunidade Inata/fisiologia , Ativação de Neutrófilo/fisiologia , Neutrófilos/fisiologia , Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Catalase/antagonistas & inibidores , Catalase/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Morte Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Sobrevivência Celular/fisiologia , Cromatina/metabolismo , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/ultraestrutura , Inibidores Enzimáticos/farmacologia , Doença Granulomatosa Crônica/metabolismo , Doença Granulomatosa Crônica/patologia , Humanos , Peróxido de Hidrogênio/farmacologia , Elastase de Leucócito/metabolismo , Microscopia Eletrônica , NADPH Oxidases/antagonistas & inibidores , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/microbiologia , Membrana Nuclear/metabolismo , Membrana Nuclear/ultraestrutura , Oniocompostos/farmacologia , Fagocitose/imunologia , Espécies Reativas de Oxigênio/metabolismo , Staphylococcus aureus/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Vacúolos/metabolismo , Vacúolos/ultraestrutura , Receptor fas/imunologia
16.
Nat Commun ; 13(1): 1030, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35210413

RESUMO

Coinfections with pathogenic microbes continually confront cervical mucosa, yet their implications in pathogenesis remain unclear. Lack of in-vitro models recapitulating cervical epithelium has been a bottleneck to study coinfections. Using patient-derived ectocervical organoids, we systematically modeled individual and coinfection dynamics of Human papillomavirus (HPV)16 E6E7 and Chlamydia, associated with carcinogenesis. The ectocervical stem cells were genetically manipulated to introduce E6E7 oncogenes to mimic HPV16 integration. Organoids from these stem cells develop the characteristics of precancerous lesions while retaining the self-renewal capacity and organize into mature stratified epithelium similar to healthy organoids. HPV16 E6E7 interferes with Chlamydia development and induces persistence. Unique transcriptional and post-translational responses induced by Chlamydia and HPV lead to distinct reprogramming of host cell processes. Strikingly, Chlamydia impedes HPV-induced mechanisms that maintain cellular and genome integrity, including mismatch repair in the stem cells. Together, our study employing organoids demonstrates the hazard of multiple infections and the unique cellular microenvironment they create, potentially contributing to neoplastic progression.


Assuntos
Chlamydia , Coinfecção , Infecções por Papillomavirus , Neoplasias do Colo do Útero , Reprogramação Celular/genética , Feminino , Papillomavirus Humano 16/genética , Humanos , Organoides , Microambiente Tumoral , Neoplasias do Colo do Útero/genética
17.
PLoS Pathog ; 5(10): e1000639, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19876394

RESUMO

Neutrophils are the first line of defense at the site of an infection. They encounter and kill microbes intracellularly upon phagocytosis or extracellularly by degranulation of antimicrobial proteins and the release of Neutrophil Extracellular Traps (NETs). NETs were shown to ensnare and kill microbes. However, their complete protein composition and the antimicrobial mechanism are not well understood. Using a proteomic approach, we identified 24 NET-associated proteins. Quantitative analysis of these proteins and high resolution electron microscopy showed that NETs consist of modified nucleosomes and a stringent selection of other proteins. In contrast to previous results, we found several NET proteins that are cytoplasmic in unstimulated neutrophils. We demonstrated that of those proteins, the antimicrobial heterodimer calprotectin is released in NETs as the major antifungal component. Absence of calprotectin in NETs resulted in complete loss of antifungal activity in vitro. Analysis of three different Candida albicans in vivo infection models indicated that NET formation is a hitherto unrecognized route of calprotectin release. By comparing wild-type and calprotectin-deficient animals we found that calprotectin is crucial for the clearance of infection. Taken together, the present investigations confirmed the antifungal activity of calprotectin in vitro and, moreover, demonstrated that it contributes to effective host defense against C. albicans in vivo. We showed for the first time that a proportion of calprotectin is bound to NETs in vitro and in vivo.


Assuntos
Candida albicans/imunologia , Complexo Antígeno L1 Leucocitário/imunologia , Neutrófilos/imunologia , Abscesso Abdominal/imunologia , Abscesso Abdominal/microbiologia , Análise de Variância , Animais , Antifúngicos/química , Antifúngicos/metabolismo , Células Cultivadas , Estruturas Celulares/química , Estruturas Celulares/imunologia , Estruturas Celulares/ultraestrutura , Histonas/química , Histonas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Imuno-Histoquímica , Complexo Antígeno L1 Leucocitário/química , Complexo Antígeno L1 Leucocitário/metabolismo , Pneumopatias Fúngicas/imunologia , Pneumopatias Fúngicas/microbiologia , Camundongos , Camundongos Knockout , Ativação de Neutrófilo
18.
Viruses ; 13(4)2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33918348

RESUMO

Bacteriophages exert strong evolutionary pressure on their microbial hosts. In their lytic lifecycle, complete bacterial subpopulations are utilized as hosts for bacteriophage replication. However, during their lysogenic lifecycle, bacteriophages can integrate into the host chromosome and alter the host's genomic make-up, possibly resulting in evolutionary important adjustments. Not surprisingly, bacteria have evolved sophisticated immune systems to protect against phage infection. Streptococcus pyogenes isolates are frequently lysogenic and their prophages have been shown to be major contributors to the virulence of this pathogen. Most S. pyogenes phage research has focused on genomic prophages in relation to virulence, but little is known about the defensive arsenal of S. pyogenes against lytic phage infection. Here, we characterized Phage A1, an S. pyogenes bacteriophage, and investigated several mechanisms that S. pyogenes utilizes to protect itself against phage predation. We show that Phage A1 belongs to the Siphoviridae family and contains a circular double-stranded DNA genome that follows a modular organization described for other streptococcal phages. After infection, the Phage A1 genome can be detected in isolated S. pyogenes survivor strains, which enables the survival of the bacterial host and Phage A1 resistance. Furthermore, we demonstrate that the type II-A CRISPR-Cas system of S. pyogenes acquires new spacers upon phage infection, which are increasingly detectable in the absence of a capsule. Lastly, we show that S. pyogenes produces membrane vesicles that bind to phages, thereby limiting the pool of phages available for infection. Altogether, this work provides novel insight into survival strategies employed by S. pyogenes to combat phage predation.


Assuntos
Viabilidade Microbiana , Fagos de Streptococcus/genética , Fagos de Streptococcus/patogenicidade , Streptococcus pyogenes/fisiologia , Streptococcus pyogenes/virologia , Sistemas CRISPR-Cas , Genoma Viral , Lisogenia , Prófagos/genética , Virulência
19.
Nat Commun ; 12(1): 6193, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702812

RESUMO

Staphylococcus aureus bi-component pore-forming leukocidins are secreted toxins that directly target and lyse immune cells. Intriguingly, one of the leukocidins, Leukocidin AB (LukAB), is found associated with the bacterial cell envelope in addition to secreted into the extracellular milieu. Here, we report that retention of LukAB on the bacterial cells provides S. aureus with a pre-synthesized active toxin that kills immune cells. On the bacteria, LukAB is distributed as discrete foci in two distinct compartments: membrane-proximal and surface-exposed. Through genetic screens, we show that a membrane lipid, lysyl-phosphatidylglycerol (LPG), and lipoteichoic acid (LTA) contribute to LukAB deposition and release. Furthermore, by studying non-covalently surface-bound proteins we discovered that the sorting of additional exoproteins, such as IsaB, Hel, ScaH, and Geh, are also controlled by LPG and LTA. Collectively, our study reveals a multistep secretion system that controls exoprotein storage and protein translocation across the S. aureus cell wall.


Assuntos
Membrana Celular/metabolismo , Parede Celular/metabolismo , Staphylococcus aureus/metabolismo , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/toxicidade , Citotoxinas/metabolismo , Citotoxinas/toxicidade , Humanos , Leucocidinas/metabolismo , Leucocidinas/toxicidade , Lipopolissacarídeos/genética , Lipopolissacarídeos/metabolismo , Lisina/genética , Lisina/metabolismo , Camundongos , Fagócitos/efeitos dos fármacos , Fosfatidilgliceróis/genética , Fosfatidilgliceróis/metabolismo , Transporte Proteico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Ácidos Teicoicos/genética , Ácidos Teicoicos/metabolismo , Fatores de Virulência/toxicidade
20.
Sci Signal ; 14(673)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33688080

RESUMO

Neutrophil extracellular traps (NETs) are structures consisting of chromatin and antimicrobial molecules that are released by neutrophils during a form of regulated cell death called NETosis. NETs trap invading pathogens, promote coagulation, and activate myeloid cells to produce type I interferons (IFNs), proinflammatory cytokines that regulate the immune system. Here, we showed that macrophages and other myeloid cells phagocytosed NETs. Once in phagosomes, NETs translocated to the cytosol, where the DNA backbones of these structures activated the innate immune sensor cyclic GMP-AMP synthase (cGAS) and induced type I IFN production. The NET-associated serine protease neutrophil elastase (NE) mediated the activation of this pathway. We showed that NET induction in mice treated with the lectin concanavalin A, a model of autoimmune hepatitis, resulted in cGAS-dependent stimulation of an IFN response, suggesting that NETs activated cGAS in vivo. Thus, our findings suggest that cGAS is a sensor of NETs, mediating immune cell activation during infection.


Assuntos
Armadilhas Extracelulares , Animais , Citosol , DNA , Camundongos , Neutrófilos , Nucleotidiltransferases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA