Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Microb Ecol ; 83(1): 216-235, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33890146

RESUMO

Invasive aspergillosis and scedosporiosis are life-threatening fungal infections with similar clinical manifestations in immunocompromised patients. Contrarily, Scedosporium apiospermum is susceptible to some azole derivative but often resistant to amphotericin B. Histopathological examination alone cannot diagnose these two fungal species. Pathogenesis studies could contribute to explore candidate protein markers for new diagnosis and treatment methods leading to a decrease in mortality. In the present study, proteomics was conducted to identify significantly altered proteins in A549 cells infected with or without Aspergillus fumigatus and S. apiospermum as measured at initial invasion. Protein validation was performed with immunogold labelling alongside immunohistochemical techniques in infected A549 cells and lungs from murine models. Further, cytokine production was measured, using the Bio-Plex-Multiplex immunoassay. The cytoskeletal proteins HSPA9, PA2G4, VAT1, PSMA2, PEX1, PTGES3, KRT1, KRT9, CLIP1 and CLEC20A were mainly changed during A. fumigatus infection, while the immunologically activated proteins WNT7A, GAPDH and ANXA2 were principally altered during S. apiospermum infection. These proteins are involved in fungal internalisation and structural destruction leading to pulmonary disorders. Interleukin (IL)-21, IL-1α, IL-22, IL-2, IL-8, IL-12, IL-17A, interferon-γ and tumour necrosis factor-α were upregulated in both aspergillosis and scedosporiosis, although more predominately in the latter, in accordance with chitin synthase-1 and matrix metalloproteinase levels. Our results demonstrated that during invasion, A. fumigatus primarily altered host cellular integrity, whereas S. apiospermum chiefly induced and extensively modulated host immune responses.


Assuntos
Aspergillus fumigatus , Citoesqueleto/microbiologia , Epitélio/microbiologia , Micoses , Scedosporium , Células A549 , Animais , Humanos , Pulmão , Camundongos
2.
mBio ; 12(1)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33468693

RESUMO

Chlamydia trachomatis is a medically significant human pathogen and is an epithelial-tropic obligate intracellular parasite. Invasion of nonprofessional phagocytes represents a crucial step in the infection process and has likely promoted the evolution of a redundant mechanism and routes of entry. Like many other viral and invasive bacterial pathogens, manipulation of the host cell cytoskeleton represents a focal point in Chlamydia entry. The advent of genetic techniques in C. trachomatis, such as creation of complete gene deletions via fluorescence-reported allelic exchange mutagenesis (FRAEM), is providing important tools to unravel the contributions of bacterial factors in these complex pathways. The type III secretion chaperone Slc1 directs delivery of at least four effectors during the invasion process. Two of these, TarP and TmeA, have been associated with manipulation of actin networks and are essential for normal levels of invasion. The functions of TarP are well established, whereas TmeA is less well characterized. We leverage chlamydial genetics and proximity labeling here to provide evidence that TmeA directly targets host N-WASP to promote Arp2/3-dependent actin polymerization. Our work also shows that TmeA and TarP influence separate, yet synergistic pathways to accomplish chlamydial entry. These data further support an appreciation that a pathogen, confined by a reductionist genome, retains the ability to commit considerable resources to accomplish bottle-neck steps during the infection process.IMPORTANCE The increasing genetic tractability of Chlamydia trachomatis is accelerating the ability to characterize the unique infection biology of this obligate intracellular parasite. These efforts are leading to a greater understanding of the molecular events associated with key virulence requirements. Manipulation of the host actin cytoskeleton plays a pivotal role throughout Chlamydia infection, yet a thorough understanding of the molecular mechanisms initiating and orchestrating actin rearrangements has lagged. Our work highlights the application of genetic manipulation to address open questions regarding chlamydial invasion, a process essential to survival. We provide definitive insight regarding the role of the type III secreted effector TmeA and how that activity relates to another prominent effector, TarP. In addition, our data implicate at least one source that contributes to the functional divergence of entry mechanisms among chlamydial species.


Assuntos
Actinas/genética , Proteínas de Bactérias/genética , Chlamydia trachomatis/genética , Citoesqueleto/metabolismo , Chaperonas Moleculares/genética , Proteína Neuronal da Síndrome de Wiskott-Aldrich/genética , Proteína 2 Relacionada a Actina/genética , Proteína 2 Relacionada a Actina/metabolismo , Proteína 3 Relacionada a Actina/genética , Proteína 3 Relacionada a Actina/metabolismo , Actinas/metabolismo , Proteínas de Bactérias/metabolismo , Linhagem Celular , Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/metabolismo , Citoesqueleto/microbiologia , Citoesqueleto/ultraestrutura , Células Epiteliais/microbiologia , Regulação da Expressão Gênica , Células HeLa , Interações Hospedeiro-Patógeno/genética , Humanos , Chaperonas Moleculares/metabolismo , Polimerização , Transdução de Sinais , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
3.
J Proteome Res ; 16(1): 87-105, 2017 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-27740763

RESUMO

The effectiveness of macrophages in the response to systemic candidiasis is crucial to an effective clearance of the pathogen. The secretion of proteins, mRNAs, noncoding RNAs and lipids through extracellular vesicles (EVs) is one of the mechanisms of communication between immune cells. EVs change their cargo to mediate different responses, and may play a role in the response against infections. Thus we have undertaken the first quantitative proteomic analysis on the protein composition of THP-1 macrophage-derived EVs during the interaction with Candida albicans. This study revealed changes in EVs sizes and in protein composition, and allowed the identification and quantification of 717 proteins. Of them, 133 proteins changed their abundance due to the interaction. The differentially abundant proteins were involved in functions relating to immune response, signaling, or cytoskeletal reorganization. THP-1-derived EVs, both from control and from Candida-infected macrophages, had similar effector functions on other THP-1-differenciated macrophages, activating ERK and p38 kinases, and increasing both the secretion of proinflammatory cytokines and the candidacidal activity; while in THP-1 nondifferenciated monocytes, only EVs from infected macrophages increased significantly the TNF-α secretion. Our findings provide new information on the role of macrophage-derived EVs in response to C. albicans infection and in macrophages communication.


Assuntos
Candida albicans/patogenicidade , Vesículas Extracelulares/imunologia , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/imunologia , Proteoma/imunologia , Candida albicans/crescimento & desenvolvimento , Diferenciação Celular , Linhagem Celular , Biologia Computacional , Citocinas/genética , Citocinas/imunologia , Citoesqueleto/imunologia , Citoesqueleto/microbiologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/imunologia , Vesículas Extracelulares/química , Vesículas Extracelulares/microbiologia , Regulação da Expressão Gênica/imunologia , Ontologia Genética , Humanos , Macrófagos/microbiologia , Anotação de Sequência Molecular , Proteoma/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
4.
PLoS One ; 10(6): e0128301, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26029930

RESUMO

Interferon-gamma (Ifnγ), a key macrophage activating cytokine, plays pleiotropic roles in host immunity. In this study, the ability of Ifnγ to induce the aggregation of resident mouse adherent peritoneal exudate cells (APECs), consisting primarily of macrophages, was investigated. Cell-cell interactions involve adhesion molecules and, upon addition of Ifnγ, CD11b re-localizes preferentially to the sites of interaction on APECs. A functional role of CD11b in enhancing aggregation is demonstrated using Reopro, a blocking reagent, and siRNA to Cd11b. Studies with NG-methyl-L-arginine (LNMA), an inhibitor of Nitric oxide synthase (Nos), NO donors, e.g., S-nitroso-N-acetyl-DL-penicillamine (SNAP) or Diethylenetriamine/nitric oxide adduct (DETA/NO), and Nos2-/- mice identified Nitric oxide (NO) induced by Ifnγ as a key regulator of aggregation of APECs. Further studies with Nos2-/- APECs revealed that some Ifnγ responses are independent of NO: induction of MHC class II and CD80. On the other hand, Nos2 derived NO is important for other functions: motility, phagocytosis, morphology and aggregation. Studies with cytoskeleton depolymerizing agents revealed that Ifnγ and NO mediate the cortical stabilization of Actin and Tubulin which contribute to aggregation of APECs. The biological relevance of aggregation of APECs was delineated using infection experiments with Salmonella Typhimurium (S. Typhimurium). APECs from orally infected, but not uninfected, mice produce high amounts of NO and aggregate upon ex vivo culture in a Nos2-dependent manner. Importantly, aggregated APECs induced by Ifnγ contain fewer intracellular S. Typhimurium compared to their single counterparts post infection. Further experiments with LNMA or Reopro revealed that both NO and CD11b are important for aggregation; in addition, NO is bactericidal. Overall, this study elucidates novel roles for Ifnγ and Nos2 in regulating Actin, Tubulin, CD11b, motility and morphology during the aggregation response of APECs. The implications of aggregation or "group behavior" of APECs are discussed in the context of host resistance to infectious organisms.


Assuntos
Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interferon gama/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Cavidade Peritoneal/citologia , Salmonella typhimurium/fisiologia , Actinas/metabolismo , Animais , Antígeno CD11b/metabolismo , Adesão Celular/efeitos dos fármacos , Agregação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Citoesqueleto/microbiologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/deficiência , Cavidade Peritoneal/microbiologia , Estabilidade Proteica/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
5.
Infect Immun ; 81(9): 3139-47, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23774597

RESUMO

Cryptococcus spp. cause fungal meningitis, a life-threatening infection that occurs predominately in immunocompromised individuals. In order for Cryptococcus neoformans to invade the central nervous system (CNS), it must first penetrate the brain endothelium, also known as the blood-brain barrier (BBB). Despite the importance of the interrelation between C. neoformans and the brain endothelium in establishing CNS infection, very little is known about this microenvironment. Here we sought to resolve the cellular and molecular basis that defines the fungal-BBB interface during cryptococcal attachment to, and internalization by, the human brain endothelium. In order to accomplish this by a systems-wide approach, the proteomic profile of human brain endothelial cells challenged with C. neoformans was resolved using a label-free differential quantitative mass spectrometry method known as spectral counting (SC). Here, we demonstrate that as brain endothelial cells associate with, and internalize, cryptococci, they upregulate the expression of several proteins involved with cytoskeleton, metabolism, signaling, and inflammation, suggesting that they are actively signaling and undergoing cytoskeleton remodeling via annexin A2, S100A10, transgelin, and myosin. Transmission electronic microscopy (TEM) analysis demonstrates dramatic structural changes in nuclei, mitochondria, the endoplasmic reticulum (ER), and the plasma membrane that are indicative of cell stress and cell damage. The translocation of HMGB1, a marker of cell injury, the downregulation of proteins that function in transcription, energy production, protein processing, and the upregulation of cyclophilin A further support the notion that C. neoformans elicits changes in brain endothelial cells that facilitate the migration of cryptococci across the BBB and ultimately induce endothelial cell necrosis.


Assuntos
Encéfalo/metabolismo , Encéfalo/microbiologia , Criptococose/microbiologia , Cryptococcus neoformans/fisiologia , Anexina A2/genética , Anexina A2/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/microbiologia , Encéfalo/citologia , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Células Cultivadas , Criptococose/genética , Criptococose/metabolismo , Cryptococcus neoformans/metabolismo , Ciclofilina A/genética , Ciclofilina A/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Citoesqueleto/microbiologia , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/microbiologia , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miosinas/genética , Miosinas/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo , Transdução de Sinais/genética , Regulação para Cima
6.
BMC Microbiol ; 12: 36, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22424399

RESUMO

BACKGROUND: The spectrin cytoskeleton is emerging as an important host cell target of enteric bacterial pathogens. Recent studies have identified a crucial role for spectrin and its associated proteins during key pathogenic processes of Listeria monocytogenes and Salmonella Typhimurium infections. Here we investigate the involvement of spectrin cytoskeletal components during the pathogenesis of the invasive pathogen Shigella flexneri. RESULTS: Immunofluorescent microscopy reveals that protein 4.1 (p4.1), but not adducin or spectrin, is robustly recruited to sites of S. flexneri membrane ruffling during epithelial cell invasion. Through siRNA-mediated knockdowns, we identify an important role for spectrin and the associated proteins adducin and p4.1 during S. flexneri invasion. Following internalization, all three proteins are recruited to the internalized bacteria, however upon generation of actin-rich comet tails, we observed spectrin recruitment to those structures in the absence of adducin or p4.1. CONCLUSION: These findings highlight the importance of the spectrin cytoskeletal network during S. flexneri pathogenesis and further demonstrate that pathogenic events that were once thought to exclusively recruit the actin cytoskeletal system require additional cytoskeletal networks.


Assuntos
Citoesqueleto/metabolismo , Disenteria Bacilar/metabolismo , Shigella flexneri/fisiologia , Espectrina/metabolismo , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/microbiologia , Disenteria Bacilar/microbiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células HeLa , Humanos
7.
J Cell Sci ; 122(Pt 2): 256-67, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19118218

RESUMO

By interacting with components of the human host, including extracellular matrix (ECM) proteins, Streptococcus pneumoniae has evolved various strategies for colonization. Here, we characterized the interaction of pneumococci with the adhesive glycoprotein vitronectin and the contribution of this protein to pneumococcal uptake by host cells in an integrin-dependent manner. Specific interaction of S. pneumoniae with the heparin-binding sites of purified multimeric vitronectin was demonstrated by flow cytometry analysis. Host-cell-bound vitronectin promoted pneumococcal adherence to and invasion into human epithelial and endothelial cells. Pneumococci were trapped by microspike-like structures, which were induced upon contact of pneumococci with host-cell-bound vitronectin. Alphavbeta3 integrin was identified as the major cellular receptor for vitronectin-mediated adherence and uptake of pneumococci. Ingestion of pneumococci by host cells via vitronectin required a dynamic actin cytoskeleton and was dependent on integrin-linked kinase (ILK), phosphatidylinositol 3-kinase (PI3K), and protein kinase B (Akt), as demonstrated by gene silencing or in inhibition experiments. In conclusion, pneumococci exploit the vitronectin-alphavbeta3-integrin complex as a cellular receptor for invasion and this integrin-mediated internalization requires the cooperation between the host signalling molecules ILK, PI3K and Akt.


Assuntos
Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Infecções Pneumocócicas/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Streptococcus pneumoniae/metabolismo , Vitronectina/metabolismo , Actinas/metabolismo , Linhagem Celular , Citoesqueleto/metabolismo , Citoesqueleto/microbiologia , Células Endoteliais/citologia , Células Endoteliais/microbiologia , Células Epiteliais/citologia , Células Epiteliais/microbiologia , Heparina/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Infecções Pneumocócicas/microbiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Can J Microbiol ; 54(9): 748-57, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18772938

RESUMO

Enteropathogenic Escherichia coli (EPEC) colonizes the intestinal mucosa and causes a cell lesion known as attachment and effacement (A/E) lesion. The molecular mechanisms for A/E lesions include injection of Tir, which is a receptor for an adhesin named intimin. The Tir-intimin interaction causes rearrangement of the cytoskeleton forming actin-rich structures called pedestals. Unfortunately, the formation of the A/E lesions and the dynamics of the actin cytoskeleton during this rearrangement induced by EPEC cannot be studied in the natural host. However, there are EPEC strains that infect rabbit (REPEC) that are genetically and pathologically similar to EPEC. Here, we used REPEC for the infection of rabbit kidney epithelial cells, line RK13, as a model to understand the actin cytoskeleton dynamics during pedestal formation. Actin-rich pedestal formation during the infection of RK13 cells by REPEC was analyzed by electron and confocal microscopy. The kinetics of infection along with the use of antibiotics for eliminating the bacteria, as well as reinfection, evidenced the plasticity of the actin cytoskeleton during pedestal formation. Thus, this model is a helpful tool for studying the dynamics of actin cytoskeleton and for correlating the data with those observed in in vivo models in rabbits experimentally infected with REPEC.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Escherichia coli Enteropatogênica/fisiologia , Células Epiteliais/metabolismo , Infecções por Escherichia coli/metabolismo , Modelos Biológicos , Actinas/química , Actinas/ultraestrutura , Animais , Linhagem Celular , Citoesqueleto/química , Citoesqueleto/microbiologia , Citoesqueleto/ultraestrutura , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/química , Células Epiteliais/microbiologia , Células Epiteliais/ultraestrutura , Infecções por Escherichia coli/microbiologia , Humanos , Coelhos
9.
PLoS Pathog ; 4(3): e1000031, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18369477

RESUMO

Internalization of the pathogenic bacterium Pseudomonas aeruginosa by non-phagocytic cells is promoted by rearrangements of the actin cytoskeleton, but the host pathways usurped by this bacterium are not clearly understood. We used RNAi-mediated gene inactivation of approximately 80 genes known to regulate the actin cytoskeleton in Drosophila S2 cells to identify host molecules essential for entry of P. aeruginosa. This work revealed Abl tyrosine kinase, the adaptor protein Crk, the small GTPases Rac1 and Cdc42, and p21-activated kinase as components of a host signaling pathway that leads to internalization of P. aeruginosa. Using a variety of complementary approaches, we validated the role of this pathway in mammalian cells. Remarkably, ExoS and ExoT, type III secreted toxins of P. aeruginosa, target this pathway by interfering with GTPase function and, in the case of ExoT, by abrogating P. aeruginosa-induced Abl-dependent Crk phosphorylation. Altogether, this work reveals that P. aeruginosa utilizes the Abl pathway for entering host cells and reveals unexpected complexity by which the P. aeruginosa type III secretion system modulates this internalization pathway. Our results furthermore demonstrate the applicability of using RNAi screens to identify host signaling cascades usurped by microbial pathogens that may be potential targets for novel therapies directed against treatment of antibiotic-resistant infections.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Macrófagos/microbiologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pseudomonas aeruginosa/patogenicidade , RNA Interferente Pequeno , ADP Ribose Transferases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Toxinas Bacterianas/metabolismo , Linhagem Celular , Citoesqueleto/microbiologia , Citoesqueleto/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Proteínas Ativadoras de GTPase/metabolismo , Inativação Gênica , Macrófagos/enzimologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-abl/genética , Pseudomonas aeruginosa/enzimologia , Interferência de RNA
10.
Adv Exp Med Biol ; 644: 158-67, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19209821

RESUMO

Ulcerative colitis (UC) is a form of chronic inflammatory bowel disease (IBD) that almost always affects the rectal mucosa and variable length of the colon in continuity and at times mucosa of the entire colon. It is not caused by any specific pathogen. Genetics, environmental factors and altered immune responses to dietary macromolecules, colonic bacteria and cellular proteins have been implicated in the pathogenesis of UC. Autoimmune response against cytoskeletal, microfilament protein tropomyosin (Tm) seems to play an important role in the pathogenesis of UC. The predominant colonic epithelial Tm isoform, hTm5, can induce both humoral (B-cells) and cellular (T-cells) response in patients with UC. Such responses are not seen in normal subjects and disease control subjects, such as patients with Crohn's disease (CD, another type of IBD) and patients with lupus. A novel observation that hTm5 is expressed on colon epithelial cell surface but not on small intestinal epithelial cells provides evidence for presentation to immune effector cells. This surface expression of hTm5 seems to be facilitated by a colon epithelial cell membrane associated protein, CEP, that acts as a chaperone for the trans-migration of hTm5 to the surface and both hTm5 and CEP are then released outside the cell. Both CEP and hTm5 expression are increased with pro-inflammatory cytokine, such as gamma-interferon. hTm5 expression in UC mucosa is also significantly increased compared to normal. Finally, autoantibodies against hTm5 observed both in circulation and in the colon mucosa of patients with UC are pathogenic causing colon epithelial cell destruction by antibody and complement mediated cytolysis.


Assuntos
Colite Ulcerativa/metabolismo , Tropomiosina/fisiologia , Animais , Autoantígenos/química , Autoimunidade , Colo/citologia , Citocinas/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/microbiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Saúde da Família , Humanos , Sistema Imunitário , Inflamação , Modelos Biológicos , Tropomiosina/metabolismo
11.
Infect Immun ; 74(1): 192-201, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16368973

RESUMO

Substantial data implicate the commensal flora as triggers for the initiation of enteric inflammation or inflammatory disease relapse. We have shown that enteric epithelia under metabolic stress respond to nonpathogenic bacteria by increases in epithelial paracellular permeability and bacterial translocation. Here we assessed the structural basis of these findings. Confluent filter-grown monolayers of the human colonic T84 epithelial cell line were treated with 0.1 mM dinitrophenol (which uncouples oxidative phosphorylation) and noninvasive, nonpathogenic Escherichia coli (strain HB101, 10(6) CFU) with or without pretreatment with various pharmacological agents. At 24 h later, apoptosis, tight-junction protein expression, transepithelial resistance (TER; a marker of paracellular permeability), and bacterial internalization and translocation were assessed. Treatment with stabilizers of microtubules (i.e., colchicine), microfilaments (i.e., jasplakinolide) and clathrin-coated pit endocytosis (i.e., phenylarsine oxide) all failed to block DNP+E. coli HB101-induced reductions in TER but effectively prevented bacterial internalization and translocation. Neither the TER defect nor the enhanced bacterial translocations were a consequence of increased apoptosis. These data show that epithelial paracellular and transcellular (i.e., bacterial internalization) permeation pathways are controlled by different mechanisms. Thus, epithelia under metabolic stress increase their endocytotic activity that can result in a microtubule-, microfilament-dependent internalization and transcytosis of bacteria. We speculate that similar events in vivo would allow excess unprocessed antigen and bacteria into the mucosa and could evoke an inflammatory response by, for example, the activation of resident or recruited immune cells.


Assuntos
Translocação Bacteriana/imunologia , Citoesqueleto/imunologia , Citoesqueleto/metabolismo , Enterócitos/metabolismo , Escherichia coli/metabolismo , Mucosa Intestinal/microbiologia , Estresse Fisiológico/metabolismo , Translocação Bacteriana/efeitos dos fármacos , Linhagem Celular Tumoral , Cloranfenicol/farmacologia , Citoesqueleto/microbiologia , Dinitrofenóis/farmacologia , Endocitose/efeitos dos fármacos , Endocitose/imunologia , Enterócitos/efeitos dos fármacos , Enterócitos/microbiologia , Escherichia coli/efeitos dos fármacos , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Estresse Fisiológico/microbiologia
12.
Infect Immun ; 74(1): 703-10, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16369027

RESUMO

Porphyromonas gingivalis, an important periodontal pathogen, is an effective colonizer of oral tissues. The organism successfully invades, multiplies in, and survives for extended periods in primary gingival epithelial cells (GECs). It is unknown whether P. gingivalis resides in the cytoplasm of infected cells throughout the infection or can spread to adjacent cells over time. We developed a technique based on flow cytofluorometry and fluorescence microscopy to study propagation of the organism at different stages of infection of GECs. Results showed that P. gingivalis spreads cell to cell and that the amount of spreading increases gradually over time. There was a very low level of propagation of bacteria to uninfected cells early in the infection (3 h postinfection), but there were 20-fold and 45-fold increases in the propagation rate after 24 h and 48 h, respectively, of infection. Immunofluorescence microscopy of infected cells suggested that intercellular translocation of P. gingivalis may be mediated through actin-based membrane protrusions, bypassing the need for release of bacteria into extracellular medium. Consistent with these observations, cytochalasin D treatment of infected cells resulted in significant inhibition of bacterial spreading. This study shows for the first time that P. gingivalis disseminates from cell to cell without passing through the extracellular space. This mechanism of spreading may allow P. gingivalis to colonize oral tissues without exposure to the humoral immune response.


Assuntos
Células Epiteliais/microbiologia , Líquido Extracelular/microbiologia , Gengiva/microbiologia , Porphyromonas gingivalis , Actinas/metabolismo , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/transmissão , Células Cultivadas , Citoesqueleto/microbiologia , Células Epiteliais/metabolismo , Citometria de Fluxo , Gengiva/citologia , Gengiva/metabolismo , Humanos , Cinética , Microscopia de Fluorescência
13.
Microbes Infect ; 6(10): 882-91, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15310464

RESUMO

Paracoccidioides brasiliensis (Pb) yeast cells can enter mammalian cells and probably manipulate the host cell environment to favor their own growth and survival. We studied the uptake of strain Pb 18 into A549 lung and Vero epithelial cells, with an emphasis on the repercussions in the cytoskeleton and the apoptosis of host cells. Cytoskeleton components of the host cells, such as actin and tubulin, were involved in the P. brasiliensis invasion process. Cytochalasin D and colchicine treatment substantially reduced invasion, indicating the functional participation of microfilaments (MFs) and microtubules (MTs) in this mechanism. Cytokeratin could also play a role in the P. brasiliensis interaction with the host. Gp43 was recognized by anti-actin and anti-cytokeratin antibodies, but not by anti-tubulin. The apoptosis induced by this fungus in infected epithelial cells was demonstrated by various techniques: TUNEL, DNA fragmentation and Bak and Bcl-2 immunocytochemical expression. DNA fragmentation was observed in infected cells but not in uninfected ones, by both TUNEL and gel electrophoresis methods. Moreover, Bcl-2 and Bak did not show any differences until 24 h after infection of cells, suggesting a competitive mechanism that allows persistence of infection. Overexpression of Bak was observed after 48 h, indicating the loss of competition between death and survival signals. In conclusion, the mechanisms of invasion of host cells, persistence within them, and the subsequent induction of apoptosis of such cells may explain the efficient dissemination of P. brasiliensis.


Assuntos
Apoptose/fisiologia , Citoesqueleto/microbiologia , Paracoccidioides/fisiologia , Paracoccidioidomicose/microbiologia , Animais , Antifúngicos/farmacologia , Aderência Bacteriana/fisiologia , Western Blotting , Chlorocebus aethiops , Colchicina/farmacologia , Citocalasina D/farmacologia , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Humanos , Cetoconazol/farmacologia , Microscopia de Fluorescência , Paracoccidioidomicose/metabolismo , Paracoccidioidomicose/patologia , Células Vero
14.
J Med Microbiol ; 52(Pt 10): 861-867, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12972579

RESUMO

Helicobacter pylori causes host epithelial cell cytoskeletal rearrangements mediated by the translocation and tyrosine phosphorylation of an outer-membrane protein, CagA, and by the vacuolating cytotoxin, VacA. However, the mechanisms by which H. pylori mediates cytoskeletal rearrangements in infected host cells need to be more clearly defined. The aim of this study was to determine the effects of H. pylori isolates from children on the architecture of host gastric epithelial cells. Gastric epithelial (AGS) cells were infected with type I (cagE(+), cagA(+), VacA(+)) H. pylori, a type II H. pylori strain (cagE(-), cagA(-), VacA(-)) or a cagE isogenic mutant. Double-labelled immune fluorescence was used to detect adherent H. pylori and the distribution of F-actin, alpha-actinin and Arp3. Both type I and type II H. pylori strains induced stress fibres in gastric epithelial cells that were not observed in uninfected cells. Type I H. pylori also induced cell elongation (hummingbird phenotype) after 4 h of infection, whereas the type II H. pylori strain did not. Less elongation occurred when AGS cells were exposed to a cagE isogenic mutant, compared with the parental strain. Confocal microscopy showed Arp3 accumulation in AGS cells infected with wild-type H. pylori, but not in response to infection with the cagE mutant. These findings indicate that type I H. pylori induce a stress fibre-like phenotype in infected gastric epithelia by a mechanism that is different from the induction of host-cell elongation. In addition to CagA and VacA, cagE also impacts on the morphology of infected gastric epithelial cells.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/microbiologia , Citoesqueleto/patologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/patologia , Helicobacter pylori/fisiologia , Proteína 3 Relacionada a Actina , Aderência Bacteriana/fisiologia , Linhagem Celular Tumoral , Criança , Citoesqueleto/metabolismo , Úlcera Duodenal/metabolismo , Úlcera Duodenal/microbiologia , Úlcera Duodenal/patologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Proteínas Nucleares/metabolismo , Fatores de Virulência/metabolismo
15.
Mol Microbiol ; 44(4): 1095-1107, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12046591

RESUMO

Enteropathogenic Escherichia coli (EPEC) is a major cause of paediatric diarrhoea and a model for the family of attaching and effacing (A/E) pathogens. A/E pathogens encode a type III secretion system to transfer effector proteins into host cells. The EPEC Tir effector protein acts as a receptor for the bacterial surface protein intimin and is involved in the formation of Cdc42-independent, actin-rich pedestal structures beneath the adhered bacteria. In this paper, we demonstrate that EPEC binding to HeLa cells also induces Tir-independent, cytoskeletal rearrangement evidenced by the early, transient formation of filopodia-like structures at sites of infection. Filopodia formation is dependent on expression of the EPEC Map effector molecule - a protein that targets mitochondria and induces their dysfunction. We show that Map-induced filopodia formation is independent of mitochondrial targeting and is abolished by cellular expression of the Cdc42 inhibitory WASP-CRIB domain, demonstrating that Map has at least two distinct functions in host cells. The transient nature of the filopodia is related to an ability of EPEC to downregulate Map-induced cell signalling that, like pedestal formation, was dependent on both Tir and intimin proteins. The ability of Tir to downregulate filopodia was impaired by disrupting a putative GTPase-activating protein (GAP) motif, suggesting that Tir may possess such a function, with its interaction with intimin triggering this activity. Furthermore, we also found that Map-induced cell signalling inhibits pedestal formation, revealing that the cellular effects of Tir and Map must be co-ordinately regulated during infection. Possible implications of the multifunctional nature of EPEC effector molecules in pathogenesis are discussed.


Assuntos
Proteínas de Escherichia coli , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Transdução de Sinais , Actinas/metabolismo , Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Proteínas de Transporte/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/microbiologia , Diarreia/metabolismo , Diarreia/microbiologia , Regulação para Baixo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Escherichia coli/genética , Escherichia coli/ultraestrutura , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Células HeLa , Humanos , Modelos Biológicos , Ligação Proteica , Pseudópodes/fisiologia , Receptores de Superfície Celular/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo
16.
Gut ; 50 Suppl 3: III9-14, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11953326

RESUMO

Bacterial enteric infections exact a heavy toll on the human population, particularly among children. Despite the explosion of knowledge on the pathogenesis of enteric diseases experienced during the past decade, the number of diarrhoeal episodes and human deaths reported worldwide remains of apocalyptic dimensions. However, our better understanding of the pathogenic mechanisms involved in the onset of diarrhoea is finally leading to preventive interventions, such as the development of enteric vaccines, that may have a significant impact on the magnitude of this human plague. The application of a multidisciplinary approach to study bacterial pathogenesis, along with the recent sequencing of entire microbial genomes, have made possible discoveries that are changing the way scientists view the bacterium-host interaction. Today, research on the molecular basis of the pathogenesis of infective diarrhoeal diseases of necessity transcends established boundaries between microbiology, cell biology, intestinal pathophysiology, and immunology. This review focuses on the most recent outcomes of this multidisciplinary effort.


Assuntos
Infecções Bacterianas/metabolismo , Diarreia/microbiologia , Enterócitos/metabolismo , Enterotoxinas/metabolismo , Transdução de Sinais , Actinas/metabolismo , Infecções Bacterianas/imunologia , Infecções Bacterianas/patologia , Translocação Bacteriana , Cálcio/metabolismo , AMP Cíclico/metabolismo , Citoesqueleto/microbiologia , Diarreia/imunologia , Enterócitos/imunologia , Enterotoxinas/genética , Humanos , Óxido Nítrico/metabolismo , Biossíntese de Proteínas , Junções Íntimas/microbiologia
17.
Dig Dis Sci ; 45(9): 1774-80, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11052319

RESUMO

The relationship between Helicobacter pylori adherence, cytotoxin production, and modification of the cytoskeletal structure was investigated by studying the effects of 12 H. pylori strains cocultured with Hep-2 epithelial cells. Bacterial strains were isolated from patients with peptic ulcer disease or nonulcer dyspepsia. Presence of the cag pathogenicity island and vacA subtypes of the strains were determined as was the production of vacuolating cytotoxin. We found that cytoskeletal rearrangements, as observed by confocal microscopy after double staining of the bacteria and the cell actin with Texas red and fluorescein-conjugated phalloidin, respectively, occurred essentially when the strains were cytotoxin producers and that the supernatants alone could also lead to these modifications.


Assuntos
Antígenos de Bactérias , Citoesqueleto/ultraestrutura , Citotoxinas/biossíntese , Células Epiteliais/ultraestrutura , Helicobacter pylori/fisiologia , Aderência Bacteriana , Proteínas de Bactérias/análise , Linhagem Celular Transformada , Citoesqueleto/microbiologia , Células Epiteliais/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Humanos , Microscopia Confocal , Vacúolos
18.
Proc Natl Acad Sci U S A ; 97(16): 8754-61, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10922031

RESUMO

Salmonella spp. have evolved the ability to enter into cells that are normally nonphagocytic. The internalization process is the result of a remarkable interaction between the bacteria and the host cells. Immediately on contact, Salmonella delivers a number of bacterial effector proteins into the host cell cytosol through the function of a specialized organelle termed the type III secretion system. Initially, two of the delivered proteins, SopE and SopB, stimulate the small GTP-binding proteins Cdc42 and Rac. SopE is an exchange factor for these GTPases, and SopB is an inositol polyphosphate phosphatase. Stimulation of Cdc42 and Rac leads to marked actin cytoskeleton rearrangements, which are further enhanced by SipA, a Salmonella protein also delivered into the host cell by the type III secretion system. SipA lowers the critical concentration of G-actin, stabilizes F-actin at the site of bacterial entry, and increases the bundling activity of the host-cell protein T-plastin (fimbrin). The cellular responses stimulated by Salmonella are short-lived; therefore, immediately after bacterial entry, the cell regains its normal architecture. Remarkably, this process is mediated by SptP, another target of the type III secretion system. SptP exert its function by serving as a GTPase-activating protein for Cdc42 and Rac, turning these G proteins off after their stimulation by the bacterial effectors SopE and SopB. The balanced interaction of Salmonella with host cells constitutes a remarkable example of the sophisticated nature of a pathogen/host relationship shaped by evolution through a longstanding coexistence.


Assuntos
Actinas/metabolismo , Citoesqueleto/microbiologia , Salmonella/fisiologia , Citoesqueleto/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Transdução de Sinais
19.
Microbes Infect ; 1(8): 621-32, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10611739

RESUMO

Some periodontal pathogens disrupt epithelial barriers and cellular adhesion to the extracellular matrix, which affects the cytoskeleton. Porphyromonas gingivalis and Actinobacillus actinomycetemcomitans exploit the cytoskeleton during their uptake by epithelial cells. Treponema denticola perturbs actin and actin-regulating pathways in host cells. Cytoskeletal dysfunction due to pathogenic bacteria may impair physiologic remodeling and wound repair in the periodontium.


Assuntos
Citoesqueleto/microbiologia , Citoesqueleto/patologia , Doenças Periodontais/microbiologia , Doenças Periodontais/patologia , Aggregatibacter actinomycetemcomitans/patogenicidade , Aggregatibacter actinomycetemcomitans/fisiologia , Citoesqueleto/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Fibroblastos/microbiologia , Fibroblastos/patologia , Homeostase , Humanos , Porphyromonas gingivalis/patogenicidade , Porphyromonas gingivalis/fisiologia , Treponema/patogenicidade , Treponema/fisiologia
20.
Mol Microbiol ; 31(3): 859-70, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10048029

RESUMO

A clonal variant of serotype M1 group A streptococcus (designated M1inv+) has been linked to severe and invasive infections, including sepsis, necrotizing fasciitis and toxic shock. High frequency internalization of cultured epithelial cells by the M1inv+ strain 90-226 is dependent upon the M1 protein. Invasion of HeLa cells was blocked by an anti-M1 antibody, invasion by an M1- strain (90-226 emm1::km) was greatly reduced, and latex beads bound to M1 protein were readily internalized by HeLa cells. Beads coated with a truncated M1 protein were internalized far less frequently. Scanning electron microscopy indicated that streptococci invade by a zipper-like mechanism, that may be mediated by interactions with host cell microvilli. Initially, internalized streptococci and streptococci undergoing endocytosis are associated with polymerized actin. Later in the internalization process, streptococcal-containing vacuoles are associated with the lysosomal membrane glycoprotein, LAMP-1.


Assuntos
Adesinas Bacterianas , Antígenos de Bactérias , Proteínas da Membrana Bacteriana Externa , Células Epiteliais/microbiologia , Streptococcus pyogenes/patogenicidade , Actinas/análise , Proteínas de Bactérias/sangue , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/fisiologia , Northern Blotting , Southern Blotting , Western Blotting , Proteínas de Transporte/sangue , Proteínas de Transporte/isolamento & purificação , Proteínas de Transporte/fisiologia , Adesão Celular , Citoesqueleto/microbiologia , Citoesqueleto/ultraestrutura , Endocitose/fisiologia , Imunofluorescência , Células HeLa/microbiologia , Células HeLa/ultraestrutura , Humanos , Microscopia Eletrônica de Varredura , Microesferas , Mutagênese , Streptococcus pyogenes/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA