Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 147(6): 1355-68, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22153078

RESUMO

Antigen (Ag) crosspresentation by dendritic cells (DCs) involves the presentation of internalized Ags on MHC class I molecules to initiate CD8+ T cell-mediated immunity in response to certain pathogens and tumor cells. Here, we identify the SNARE Sec22b as a specific regulator of Ag crosspresentation. Sec22b localizes to the ER-Golgi intermediate compartment (ERGIC) and pairs to the plasma membrane SNARE syntaxin 4, which is present in phagosomes (Phgs). Depletion of Sec22b inhibits the recruitment of ER-resident proteins to Phgs and to the vacuole containing the Toxoplasma gondii parasite. In Sec22b-deficient DCs, crosspresentation is compromised after Ag phagocytosis or endocytosis and after invasion by T. gondii. Sec22b silencing inhibited Ag export to the cytosol and increased phagosomal degradation by accelerating lysosomal recruitment. Our findings provide insight into an intracellular traffic pathway required for crosspresentation and show that Sec22b-dependent recruitment of ER proteins to Phgs critically influences phagosomal functions in DCs.


Assuntos
Apresentação de Antígeno , Células Dendríticas/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli , Fagossomos/imunologia , Proteínas R-SNARE/metabolismo , Toxoplasma , Toxoplasmose/imunologia , Animais , Reações Cruzadas , Células Dendríticas/citologia , Camundongos , Camundongos Endogâmicos C57BL
2.
Mol Microbiol ; 121(4): 636-645, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37975530

RESUMO

Bacterial secretion systems, such as the type 3, 4, and 6 are multiprotein nanomachines expressed at the surface of pathogens with Gram-negative like envelopes. They are known to be crucial for virulence and to translocate bacteria-encoded effector proteins into host cells to manipulate cellular functions. This facilitates either pathogen attachment or invasion of the targeted cell. Effector proteins also promote evasion of host immune recognition. Imaging by cryo-electron microscopy in combination with structure determination has become a powerful approach to understand how these nanomachines work. Still, questions on their assembly, the precise secretion mechanisms, and their direct involvement in pathogenicity remain unsolved. Here, we present an overview of the recent developments in in situ cryo-electron microscopy. We discuss its potential for the investigation of the role of bacterial secretion systems during the host-bacterial crosstalk at the molecular level. These in situ studies open new perspectives for our understanding of secretion system structure and function.


Assuntos
Sistemas de Secreção Bacterianos , Tomografia com Microscopia Eletrônica , Tomografia com Microscopia Eletrônica/métodos , Microscopia Crioeletrônica , Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Tipo III/metabolismo
3.
PLoS Pathog ; 17(4): e1009550, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33930101

RESUMO

Salmonella Typhimurium (S. Typhimurium) is an enteric bacterium capable of invading a wide range of hosts, including rodents and humans. It targets different host cell types showing different intracellular lifestyles. S. Typhimurium colonizes different intracellular niches and is able to either actively divide at various rates or remain dormant to persist. A comprehensive tool to determine these distinct S. Typhimurium lifestyles remains lacking. Here we developed a novel fluorescent reporter, Salmonella INtracellular Analyzer (SINA), compatible for fluorescence microscopy and flow cytometry in single-bacterium level quantification. This identified a S. Typhimurium subpopulation in infected epithelial cells that exhibits a unique phenotype in comparison to the previously documented vacuolar or cytosolic S. Typhimurium. This subpopulation entered a dormant state in a vesicular compartment distinct from the conventional Salmonella-containing vacuoles (SCV) as well as the previously reported niche of dormant S. Typhimurium in macrophages. The dormant S. Typhimurium inside enterocytes were viable and expressed Salmonella Pathogenicity Island 2 (SPI-2) virulence factors at later time points. We found that the formation of these dormant S. Typhimurium is not triggered by the loss of SPI-2 effector secretion but it is regulated by (p)ppGpp-mediated stringent response through RelA and SpoT. We predict that intraepithelial dormant S. Typhimurium represents an important pathogen niche and provides an alternative strategy for S. Typhimurium pathogenicity and its persistence.


Assuntos
Células Epiteliais/microbiologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/fisiologia , Latência Viral/fisiologia , Células 3T3 , Animais , Células CACO-2 , Células Epiteliais/patologia , Ilhas Genômicas/genética , Células HeLa , Humanos , Camundongos , Infecções por Salmonella/patologia , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidade , Células THP-1 , Vacúolos/microbiologia , Vacúolos/patologia , Fatores de Virulência/genética , Latência Viral/genética
4.
J Infect Dis ; 225(6): 1005-1010, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-32582947

RESUMO

The bacterial pathogen Listeria monocytogenes invades host cells, ruptures the internalization vacuole, and reaches the cytosol for replication. A high-content small interfering RNA (siRNA) microscopy screen allowed us to identify epithelial cell factors involved in L. monocytogenes vacuolar rupture, including the serine/threonine kinase Taok2. Kinase activity inhibition using a specific drug validated a role for Taok2 in favoring L. monocytogenes cytoplasmic access. Furthermore, we showed that Taok2 recruitment to L. monocytogenes vacuoles requires the presence of pore-forming toxin listeriolysin O. Overall, our study identified the first set of host factors modulating L. monocytogenes vacuolar rupture and cytoplasmic access in epithelial cells.


Assuntos
Listeria monocytogenes , Listeriose , Proteínas de Bactérias , Citoplasma , Citosol , Proteínas Hemolisinas , Humanos , Listeriose/microbiologia , Vacúolos/microbiologia , Vacúolos/fisiologia
5.
PLoS Pathog ; 16(8): e1008822, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32866204

RESUMO

Shigella flexneri invades host cells by entering within a bacteria-containing vacuole (BCV). In order to establish its niche in the host cytosol, the bacterium ruptures its BCV. Contacts between S. flexneri BCV and infection-associated macropinosomes (IAMs) formed in situ have been reported to enhance BCV disintegration. The mechanism underlying S. flexneri vacuolar escape remains however obscure. To decipher the molecular mechanism priming the communication between the IAMs and S. flexneri BCV, we performed mass spectrometry-based analysis of the magnetically purified IAMs from S. flexneri-infected cells. While proteins involved in host recycling and exocytic pathways were significantly enriched at the IAMs, we demonstrate more precisely that the S. flexneri type III effector protein IpgD mediates the recruitment of the exocyst to the IAMs through the Rab8/Rab11 pathway. This recruitment results in IAM clustering around S. flexneri BCV. More importantly, we reveal that IAM clustering subsequently facilitates an IAM-mediated unwrapping of the ruptured vacuole membranes from S. flexneri, enabling the naked bacterium to be ready for intercellular spread via actin-based motility. Taken together, our work untangles the molecular cascade of S. flexneri-driven host trafficking subversion at IAMs to develop its cytosolic lifestyle, a crucial step en route for infection progression at cellular and tissue level.


Assuntos
Disenteria Bacilar , Shigella flexneri , Transdução de Sinais , Vacúolos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Disenteria Bacilar/genética , Disenteria Bacilar/metabolismo , Células HeLa , Humanos , Shigella flexneri/genética , Shigella flexneri/metabolismo , Shigella flexneri/patogenicidade , Vacúolos/genética , Vacúolos/metabolismo , Vacúolos/microbiologia , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
6.
PLoS Pathog ; 16(4): e1008446, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32282860

RESUMO

Microfold (M) cell host-pathogen interaction studies would benefit from the visual analysis of dynamic cellular and microbial interplays. We adapted a human in vitro M cell model to physiological bacterial infections, expression of fluorescent localization reporters and long-term three-dimensional time-lapse microscopy. This approach allows following key steps of M cell infection dynamics at subcellular resolution, from the apical onset to basolateral epithelial dissemination. We focused on the intracellular pathogen Shigella flexneri, classically reported to transcytose through M cells to initiate bacillary dysentery in humans, while eliciting poorly protective immune responses. Our workflow was critical to reveal that S. flexneri develops a bimodal lifestyle within M cells leading to rapid transcytosis or delayed vacuolar rupture, followed by direct actin motility-based propagation to neighboring enterocytes. Moreover, we show that Listeria monocytogenes, another intracellular pathogen sharing a tropism for M cells, disseminates in a similar manner and evades M cell transcytosis completely. We established that actin-based M cell-to-enterocyte spread is the major dissemination pathway for both pathogens and avoids their exposure to basolateral compartments in our system. Our results challenge the notion that intracellular pathogens are readily transcytosed by M cells to inductive immune compartments in vivo, providing a potential mechanism for their ability to evade adaptive immunity.


Assuntos
Disenteria Bacilar/microbiologia , Enterócitos/microbiologia , Células Epiteliais/microbiologia , Listeria monocytogenes/fisiologia , Listeriose/microbiologia , Shigella flexneri/fisiologia , Células CACO-2 , Humanos , Listeria monocytogenes/genética , Shigella flexneri/genética
7.
Cell Microbiol ; 23(7): e13342, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33848057

RESUMO

Large volumes of liquid and other materials from the extracellular environment are internalised by eukaryotic cells via an endocytic process called macropinocytosis. It is now recognised that this fundamental and evolutionarily conserved pathway is hijacked by numerous intracellular pathogens as an entry portal to the host cell interior. Yet, an increasing number of additional cellular functions of macropinosomes in pathologic processes have been reported beyond this role for fluid internalisation. It emerges that the identity of macropinosomes can vary hugely and change rapidly during their lifetime. A deeper understanding of this important multi-faceted compartment is based on novel methods for their investigation. These methods are either imaging-based for the tracking of macropinosome dynamics, or they provide the means to extract macropinosomes at high purity for comprehensive proteomic analyses. Here, we portray these new approaches for the investigation of macropinosomes. We document how these method developments have provided insights for a new understanding of the intracellular lifestyle of the bacterial pathogens Shigella and Salmonella. We suggest that a systematic complete characterisation of macropinosome subversion with these approaches during other infection processes and pathologies will be highly beneficial for our understanding of the underlying cellular and molecular processes.


Assuntos
Disenteria Bacilar/microbiologia , Endossomos/microbiologia , Interações Hospedeiro-Patógeno , Infecções por Salmonella/microbiologia , Salmonella/patogenicidade , Shigella/patogenicidade , Humanos
8.
Cell Microbiol ; 23(1): e13263, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32945061

RESUMO

The ability of Salmonella to survive and replicate within mammalian host cells involves the generation of a membranous compartment known as the Salmonella-containing vacuole (SCV). Salmonella employs a number of effector proteins that are injected into host cells for SCV formation using its type-3 secretion systems encoded in SPI-1 and SPI-2 (T3SS-1 and T3SS-2, respectively). Recently, we reported that S. Typhimurium requires T3SS-1 and T3SS-2 to survive in the model amoeba Dictyostelium discoideum. Despite these findings, the involved effector proteins have not been identified yet. Therefore, we evaluated the role of two major S. Typhimurium effectors SopB and SifA during D. discoideum intracellular niche formation. First, we established that S. Typhimurium resides in a vacuolar compartment within D. discoideum. Next, we isolated SCVs from amoebae infected with wild type or the ΔsopB and ΔsifA mutant strains of S. Typhimurium, and we characterised the composition of this compartment by quantitative proteomics. This comparative analysis suggests that S. Typhimurium requires SopB and SifA to modify the SCV proteome in order to generate a suitable intracellular niche in D. discoideum. Accordingly, we observed that SopB and SifA are needed for intracellular survival of S. Typhimurium in this organism. Thus, our results provide insight into the mechanisms employed by Salmonella to survive intracellularly in phagocytic amoebae.


Assuntos
Proteínas de Bactérias/metabolismo , Dictyostelium/metabolismo , Proteoma/metabolismo , Salmonella typhimurium/metabolismo , Vacúolos/metabolismo , Amoeba/metabolismo , Animais , Proteínas de Bactérias/genética , Interações Hospedeiro-Patógeno , Mutação , Proteômica , Proteínas de Protozoários/metabolismo , Infecções por Salmonella/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/genética
9.
EMBO J ; 36(17): 2567-2580, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28701483

RESUMO

The role of second messengers in the diversion of cellular processes by pathogens remains poorly studied despite their importance. Among these, Ca2+ virtually regulates all known cell processes, including cytoskeletal reorganization, inflammation, or cell death pathways. Under physiological conditions, cytosolic Ca2+ increases are transient and oscillatory, defining the so-called Ca2+ code that links cell responses to specific Ca2+ oscillatory patterns. During cell invasion, Shigella induces atypical local and global Ca2+ signals. Here, we show that by hydrolyzing phosphatidylinositol-(4,5)bisphosphate, the Shigella type III effector IpgD dampens inositol-(1,4,5)trisphosphate (InsP3) levels. By modifying InsP3 dynamics and diffusion, IpgD favors the elicitation of long-lasting local Ca2+ signals at Shigella invasion sites and converts Shigella-induced global oscillatory responses into erratic responses with atypical dynamics and amplitude. Furthermore, IpgD eventually inhibits InsP3-dependent responses during prolonged infection kinetics. IpgD thus acts as a pathogen regulator of the Ca2+ code implicated in a versatility of cell functions. Consistent with this function, IpgD prevents the Ca2+-dependent activation of calpain, thereby preserving the integrity of cell adhesion structures during the early stages of infection.


Assuntos
Proteínas de Bactérias/metabolismo , Cálcio/metabolismo , Disenteria Bacilar/metabolismo , Interações Hospedeiro-Patógeno , Monoéster Fosfórico Hidrolases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Shigella flexneri/fisiologia , Calpaína/metabolismo , Adesão Celular , Células HeLa , Humanos , Transdução de Sinais
10.
Cell Microbiol ; 22(4): e13190, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32185894

RESUMO

The discovery of the role of ActA to polymerise actin at one pole of Listeria monocytogenes represents a key event in the field of cellular microbiology. It uncovered much more than the molecular principle behind actin-based motility of Listeria within the cytosol of infected cells, and it changed the way how actin dynamics could be studied and eventually understood. The ActA discovery took place at a time when cell biology, biochemistry and microbiology came together in a very fruitful fashion. Here, we provide an overview of the science that took place around this event. Then, we outline the wide array of research fields that have been impacted by this finding. This ranges from structural and biophysical investigations on actin and its dynamics, the role of actin polymerisation during infection with different pathogens, to actin-dynamics during various pathologies. Like a comet in the sky, Pascale Cossart's work on ActA has inspired and will inspire generations of (life) scientists.


Assuntos
Actinas/fisiologia , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Proteínas do Citoesqueleto , Citosol/microbiologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Movimento , Polimerização
11.
Genes Immun ; 20(5): 426-435, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31019256

RESUMO

Only a profound understanding of the structure and function of cells-either as single units or in the context of tissues and whole organisms-will allow a comprehension of what happens in pathological conditions and provides the means to fight disease. The Cell Biology and Infection (BCI for Biologie Cellulaire et Infection) department was created in 2002 at the Institut Pasteur in Paris to develop a research program under the umbrella of cell biology, infection biology, and microbiology. Its visionary ambition was to shape a common framework for cellular microbiology, and to interface the latter with hard sciences like physics and mathematics and cutting-edge technology. This concept, ahead of time, has given high visibility to the field of cellular microbiology and quantitative cell biology, and it has allowed the successful execution of highly interdisciplinary research programs linking a molecular understanding of cellular events with disease. Now, the BCI department embraces additional pathologies, namely cancer and neurodegenerative diseases. Here, we will portray how the integrative research approach of BCI has led to major scientific breakthroughs during the last 10 years, and where we see scientific opportunities for the near future.


Assuntos
Técnicas Citológicas/métodos , Infectologia/métodos , Pesquisa Interdisciplinar/métodos , França
12.
Cell Microbiol ; 20(4)2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29250873

RESUMO

Salmonella enterica induces membrane ruffling and genesis of macropinosomes during its interactions with epithelial cells. This is achieved through the type three secretion system-1, which first mediates bacterial attachment to host cells and then injects bacterial effector proteins to alter host behaviour. Next, Salmonella enters into the targeted cell within an early membrane-bound compartment that matures into a slow growing, replicative niche called the Salmonella Containing Vacuole (SCV). Alternatively, the pathogen disrupts the membrane of the early compartment and replicate at high rate in the cytosol. Here, we show that the in situ formed macropinosomes, which have been previously postulated to be relevant for the step of Salmonella entry, are key contributors for the formation of the mature intracellular niche of Salmonella. We first clarify the primary mode of type three secretion system-1 induced Salmonella entry into epithelial cells by combining classical fluorescent microscopy with cutting edge large volume electron microscopy. We observed that Salmonella, similarly to Shigella, enters epithelial cells inside tight vacuoles rather than in large macropinosomes. We next apply this technology to visualise rupturing Salmonella containing compartments, and we use extended time-lapse microscopy to establish early markers that define which Salmonella will eventually hyper replicate. We show that at later infection stages, SCVs harbouring replicating Salmonella have previously fused with the in situ formed macropinosomes. In contrast, such fusion events could not be observed for hyper-replicating Salmonella, suggesting that fusion of the Salmonella entry compartment with macropinosomes is the first committed step of SCV formation.


Assuntos
Células Epiteliais/microbiologia , Células Epiteliais/ultraestrutura , Infecções por Salmonella/microbiologia , Infecções por Salmonella/patologia , Salmonella enterica/fisiologia , Citosol/metabolismo , Citosol/ultraestrutura , Células HeLa , Interações Hospedeiro-Patógeno , Humanos
13.
Semin Cell Dev Biol ; 60: 155-167, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27448494

RESUMO

Bacterial pathogens display an impressive arsenal of molecular mechanisms that allow survival in diverse host niches. Subversion of plasma membrane and cytoskeletal functions are common themes associated to infection by both extracellular and intracellular pathogens. Moreover, intracellular pathogens modify the structure/stability of their membrane-bound compartments and escape degradation from phagocytic or autophagic pathways. Here, we review the manipulation of host membranes by Listeria monocytogenes, Francisella tularensis, Shigella flexneri and Yersinia spp. These four bacterial model pathogens exemplify generalized strategies as well as specific features observed during bacterial infection processes.


Assuntos
Membrana Celular/microbiologia , Francisella/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Listeria/fisiologia , Shigella/fisiologia , Yersinia/fisiologia , Animais , Humanos
14.
Infect Immun ; 86(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29084895

RESUMO

Salmonella targets and enters epithelial cells at permissive entry sites: some cells are more likely to be infected than others. However, the parameters that lead to host cell heterogeneity are not known. Here, we quantitatively characterized host cell vulnerability to Salmonella infection based on imaged parameters. We performed successive infections of the same host cell population followed by automated high-throughput microscopy and observed that infected cells have a higher probability of being reinfected. Establishing a predictive model, we identified two combined origins of host cell vulnerability: pathogen-induced cellular vulnerability emerging from Salmonella uptake and persisting at later stages of the infection and host cell-inherent vulnerability. We linked the host cell-inherent vulnerability with its morphological attributes, such as local cell crowding, and with host cell cholesterol content. This showed that the probability of Salmonella infection success can be forecast from morphological or molecular host cell parameters.


Assuntos
Salmonella typhimurium/fisiologia , Células CACO-2 , Sobrevivência Celular , Colesterol/metabolismo , Células HeLa , Humanos , Microscopia/métodos , Modelos Biológicos
15.
PLoS Pathog ; 12(5): e1005602, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27182929

RESUMO

Intracellular pathogens include all viruses, many bacteria and parasites capable of invading and surviving within host cells. Key to survival is the subversion of host cell pathways by the pathogen for the purpose of propagation and evading the immune system. The intracellular bacterium Shigella flexneri, the causative agent of bacillary dysentery, invades host cells in a vacuole that is subsequently ruptured to allow growth of the pathogen within the host cytoplasm. S. flexneri invasion has been classically described as a macropinocytosis-like process, however the underlying details and the role of macropinosomes in the intracellular bacterial lifestyle have remained elusive. We applied dynamic imaging and advanced large volume correlative light electron microscopy (CLEM) to study the highly transient events of S. flexneri's early invasion into host epithelial cells and elucidate some of its fundamental features. First, we demonstrate a clear distinction between two compartments formed during the first step of invasion: the bacterial containing vacuole and surrounding macropinosomes, often considered identical. Next, we report a functional link between macropinosomes and the process of vacuolar rupture, demonstrating that rupture timing is dependent on the availability of macropinosomes as well as the activity of the small GTPase Rab11 recruited directly to macropinosomes. We go on to reveal that the bacterial containing vacuole and macropinosomes come into direct contact at the onset of vacuolar rupture. Finally, we demonstrate that S. flexneri does not subvert pre-existing host endocytic vesicles during the invasion steps leading to vacuolar rupture, and propose that macropinosomes are the major compartment involved in these events. These results provide the basis for a new model of the early steps of S. flexneri epithelial cell invasion, establishing a different view of the enigmatic process of cytoplasmic access by invasive bacterial pathogens.


Assuntos
Disenteria Bacilar/microbiologia , Endossomos/microbiologia , Células Epiteliais/microbiologia , Shigella flexneri/patogenicidade , Vacúolos/ultraestrutura , Endossomos/ultraestrutura , Células Epiteliais/ultraestrutura , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Processamento de Imagem Assistida por Computador , Microscopia/métodos , Pinocitose/fisiologia
16.
Methods ; 127: 12-22, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28522322

RESUMO

Macropinocytosis is the uptake of extracellular fluid within vesicles of varying size that takes place during numerous cellular processes in a large variety of cells. A growing number of pathogens, including viruses, parasites, and bacteria are known to induce macropinocytosis during their entry into targeted host cells. We have recently discovered that the human enteroinvasive, bacterial pathogen Shigella causes in situ macropinosome formation during its entry into epithelial cells. These infection-associated macropinosomes are not generated to ingest the bacteria, but are instead involved in Shigella's intracellular niche formation. They make contacts with the phagocytosed shigellae to promote vacuolar membrane rupture and their cytosolic release. Here, we provide an overview of the different imaging approaches that are currently used to analyze macropinocytosis during infectious processes with a focus on Shigella entry. We detail the advantages and disadvantages of genetically encoded reporters as well as chemical probes to trace fluid phase uptake. In addition, we report how such reporters can be combined with ultrastructural approaches for correlative light electron microscopy either in thin sections or within large volumes. The combined imaging techniques introduced here provide a detailed characterization of macropinosomes during bacterial entry, which, apart from Shigella, are relevant for numerous other ones, including Salmonella, Brucella or Mycobacteria.


Assuntos
Técnicas Bacteriológicas/métodos , Disenteria Bacilar/diagnóstico por imagem , Endossomos/ultraestrutura , Interações Hospedeiro-Patógeno , Pinocitose , Biomarcadores , Disenteria Bacilar/fisiopatologia , Endossomos/microbiologia , Humanos , Microscopia Eletrônica/métodos , Shigella
17.
J Cell Sci ; 128(13): 2373-87, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25977475

RESUMO

Antigen-presenting cells have the remarkable capacity to transfer exogenous antigens to the cytosol for processing by proteasomes and subsequent presentation on major histocompatibility complex class-I (MHC-I) molecules, a process termed cross-presentation. This is the target of biomedical approaches that aim to trigger a therapeutic immune response. The receptor-binding B-subunit of Shiga toxin (STxB) has been developed as an antigen delivery tool for such immunotherapy applications. In this study, we have analyzed pathways and trafficking factors that are involved in this process. A covalent conjugate between STxB and saporin was generated to quantitatively sample the membrane translocation step to the cytosol in differentiated monocyte-derived THP-1 cells. We have found that retrograde trafficking to the Golgi complex was not required for STxB-saporin translocation to the cytosol or for STxB-dependent antigen cross-presentation. Depletion of endosomal Rab7 inhibited, and lowering membrane cholesterol levels favored STxB-saporin translocation. Interestingly, experiments with reducible and non-reducible linker-arm-STxB conjugates led to the conclusion that after translocation, STxB remains associated with the cytosolic membrane leaflet. In summary, we report new facets of the endosomal escape process bearing relevance to antigen cross-presentation.


Assuntos
Citosol/metabolismo , Toxina Shiga/metabolismo , Transporte Biológico , Linfócitos T CD8-Positivos/imunologia , Compartimento Celular , Citomegalovirus/fisiologia , Endocitose , Endossomos/metabolismo , Epitopos/metabolismo , Transferência Ressonante de Energia de Fluorescência , Células HeLa , Humanos , Biossíntese de Proteínas , Proteínas Inativadoras de Ribossomos Tipo 1/metabolismo , Saporinas , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
18.
Cell Microbiol ; 18(3): 330-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26762760

RESUMO

Invasive bacterial pathogens are engulfed upon host cell entry in a vacuolar environment called the bacteria-containing vacuole (BCV). BCVs directly contact with numerous host compartments, mainly vesicles of the endocytic pathway, such as endosomes or lysosomes. In addition, they also interact with the endoplasmic reticulum and endomembranes of the secretory pathway. These connections between the pathogen and the host occur either through heterotypic membrane fusions or through membrane contact sites. The precise regulation of BCV contacts with host compartments defines the constitution of the intracellular bacterial niche. It emerges that the associated pathways may control the stability of the BCV resulting either in vacuolar or cytoplasmically growing bacteria. Here, we will portray how the usage of novel proteomics and imaging technologies allows comparison of the communication of different host cell compartments with four relevant intracellular human pathogens, namely Salmonella enterica, Legionella pneumophila, Shigella flexneri and Francisella tularensis. The first two remain mainly within the BCV, and the latter two escape into the cytoplasm.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Organelas/microbiologia , Salmonella enterica/patogenicidade , Vacúolos/microbiologia , Francisella tularensis/patogenicidade , Humanos , Legionella pneumophila/patogenicidade , Shigella flexneri/patogenicidade
19.
Cell Microbiol ; 18(8): 1070-7, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27247079

RESUMO

Mycobacterium tuberculosis, the infectious agent of human tuberculosis is a master player in circumventing the defense mechanisms of the host immune system. The host-pathogen interaction in the case of an infection with M. tuberculosis is highly complex, involving dedicated mycobacterial virulence factors as well as the action of the innate and adapted immune systems, which determine the outcome of infection. Macrophages play a key role in this process through internalizing the bacterium in a phagosomal vacuole. While this action has normally the function of eliminating invading bacteria, M. tuberculosis employs efficient strategies to prevent its extermination. The question on how-and-where the bacterium succeeds in doing so has interested generations of scientists and still remains a fascinating and important research subject focused on mycobacterial lipids, secretion systems and other contributing factors. This topic is also central to the longstanding and partially controversial discussion on mycobacterial phagosomal rupture and vacuole-to-cytosol translocation, to be reviewed here in more detail.


Assuntos
Citosol/microbiologia , Mycobacterium tuberculosis/fisiologia , Tuberculose/microbiologia , Vacúolos/microbiologia , Animais , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Fagossomos/microbiologia , Tuberculose/imunologia
20.
Appl Environ Microbiol ; 82(9): 2700-2708, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26921426

RESUMO

Bacterial type III secretion system (T3SS) effector proteins are critical determinants of infection for many animal and plant pathogens. However, monitoring of the translocation and delivery of these important virulence determinants has proved to be technically challenging. Here, we used a genetically engineered LOV (light-oxygen-voltage) sensing domain derivative to monitor the expression, translocation, and localization of bacterial T3SS effectors. We found the Escherichia coli O157:H7 bacterial effector fusion Tir-LOV was functional following its translocation and localized to the host cell membrane in discrete foci, demonstrating that LOV-based reporters can be used to visualize the effector translocation with minimal manipulation and interference. Further evidence for the versatility of the reporter was demonstrated by fusing LOV to the C terminus of the Shigella flexneri effector IpaB. IpaB-LOV localized preferentially at bacterial poles before translocation. We observed the rapid translocation of IpaB-LOV in a T3SS-dependent manner into host cells, where it localized at the bacterial entry site within membrane ruffles.


Assuntos
Proteínas de Bactérias/metabolismo , Genes Reporter , Sistemas de Secreção Tipo III/metabolismo , Proteínas de Bactérias/genética , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Engenharia Genética/métodos , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Imagem Óptica , Domínios Proteicos , Shigella flexneri/genética , Shigella flexneri/metabolismo , Sistemas de Secreção Tipo III/análise , Sistemas de Secreção Tipo III/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA