Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anat Rec (Hoboken) ; 306(5): 941-946, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36866415

RESUMO

The gastrointestinal system is classically known for its function in digesting food for nutrient uptake, but it plays a much larger role in the general health of organisms. Understanding the relationships between the gastrointestinal tract and inflammation, the nervous system, diseases caused through disregulation of molecular components as well as its association with beneficial and pathogenic microbes have been the focus of intense research over the many decades. In this Special Issue we delve into histological, molecular, and evolutionary aspects of gastrointestinal system components in healthy and diseased tissues, to give a broad perspective on the different organs that make-up this system.


Assuntos
Trato Gastrointestinal , Trato Gastrointestinal/anatomia & histologia
2.
Anat Rec (Hoboken) ; 306(5): 1140-1148, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-35488878

RESUMO

The actin cytoskeleton forms much of the structure needed for the intracellular motility of an assortment of microbes as well as entire cells. The co-factor to the ubiquitin conjugating enzyme Ube2N (Ube2V1) has been implicated in both cancer cell metastasis and lysine-63 ubiquitylation of ß actin. As this protein complexes with Ube2N, we sought to investigate whether Ube2N itself was involved in actin-based events occurring during the Listeria monocytogenes infections as well as within motile whole cells. Through examination of L. monocytogenes actin clouds, comet tails and membrane protrusions as well as lamellipodia in migrating cells, we show that Ube2N is recruited to actin-rich structures. When pharmacologically inhibited we demonstrate that Ube2N is crucial for the function of actin-rich structures when associated with the plasma membrane.


Assuntos
Listeria monocytogenes , Listeria , Actinas/metabolismo , Listeria/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Pseudópodes/metabolismo , Listeria monocytogenes/metabolismo
3.
Anat Rec (Hoboken) ; 306(5): 1088-1110, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-35582740

RESUMO

Shigella flexneri (S. flexneri), the causative agent of bacillary dysentery, uses an effector-mediated strategy to hijack host cells and cause disease. To propagate and spread within human tissues, S. flexneri bacteria commandeer the host actin cytoskeleton to generate slender actin-rich comet tails to move intracellularly, and later, plasma membrane actin-based protrusions to move directly between adjacent host cells. To facilitate intercellular bacterial spreading, large micron-sized endocytic-like membrane invaginations form at the periphery of neighboring host cells that come into contact with S. flexneri-containing membrane protrusions. While S. flexneri comet tails and membrane protrusions consist primarily of host actin cytoskeletal proteins, S. flexneri membrane invaginations remain poorly understood with only clathrin and the clathrin adapter epsin-1 localized to the structures. Tangentially, we recently reported that Listeria monocytogenes, another actin-hijacking pathogen, exploits an assortment of caveolar and actin-bundling proteins at their micron-sized membrane invaginations formed during their cell-to-cell movement. Thus, to further characterize the S. flexneri disease process, we set out to catalog the distribution of a variety of actin-associated and caveolar proteins during S. flexneri actin-based motility and cell-to-cell spreading. Here we show that actin-associated proteins found at L. monocytogenes comet tails and membrane protrusions mimic those present at S. flexneri comet tails with the exception of α-actinins 1 and 4, which were shed from S. flexneri membrane protrusions. We also demonstrate that all known host endocytic components found at L. monocytogenes membrane invaginations are also present at those formed during S. flexneri infections.


Assuntos
Actinas , Listeria monocytogenes , Humanos , Actinas/metabolismo , Shigella flexneri/metabolismo , Movimento Celular , Listeria monocytogenes/metabolismo , Clatrina , Células HeLa
4.
Cell Microbiol ; 21(3): e12977, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30415487

RESUMO

Klebsiella pneumoniae raises significant concerns to the health care industry as these microbes are the source of widespread contamination of medical equipment, cause pneumonia as well as other multiorgan metastatic infections and have gained multidrug resistance. Despite soaring mortality rates, the host cell alterations occurring during these infections remain poorly understood. Here, we show that during in vitro and in vivo K. pneumoniae infections of lung epithelia, microtubules are severed and then eliminated. This destruction does not require direct association of K. pneumoniae with the host cells, as microtubules are disassembled in cells that are distant from the infecting bacteria. This microtubule dismantling is dependent on the K. pneumoniae (Kp) gene ytfL as non-pathogenic Escherichia coli expressing Kp ytfL disassemble microtubules in the absence of K. pneumoniae itself. Our data points to the host katanin catalytic subunit A like 1 protein (KATNAL1) and the katanin regulatory subunit B1 protein (KATNB1) as the gatekeepers to the microtubule severing event as both proteins localise specifically to microtubule cut sites. Infected cells that had either of these proteins knocked out maintained intact microtubules. Taken together, we have identified a novel mechanism that a bacterial pathogen has exploited to cause microtubule destruction within the host epithelia.


Assuntos
Proteínas de Bactérias/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Interações Hospedeiro-Patógeno , Klebsiella pneumoniae/crescimento & desenvolvimento , Microtúbulos/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Humanos , Infecções por Klebsiella/patologia , Klebsiella pneumoniae/patogenicidade , Camundongos Endogâmicos C57BL , Modelos Teóricos , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Fatores de Virulência/metabolismo
5.
Exp Cell Res ; 369(1): 139-146, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29778753

RESUMO

The host actin cytoskeleton is utilized by an assortment of pathogenic bacteria to colonize and cause disease in their hosts. Two prominently studied actin-hijacking bacteria are enteropathogenic Escherichia coli (EPEC) and Listeria monocytogenes. EPEC form actin-rich pedestals atop its host cells to move across the intestinal epithelia, while Listeria monocytogenes generate branched actin networks arranged as actin clouds around the bacteria and as comet tails for propulsion within and amongst their host cells. Previous mass spectrometry analysis revealed that a member of the calponin family of actin-bundling proteins, transgelin/SM22 was enriched in EPEC pedestals. To validate that finding and examine the role of SM22 during infections, we initially immunolocalized SM22 in EPEC and L. monocytogenes infected cells, used siRNA to deplete SM22 and EGFP-SM22 to overexpress SM22, then quantified the alterations to the bacterially generated actin structures. SM22 concentrated at all bacterially-generated actin structures. Depletion of SM22 resulted in fewer pedestals and comet tails and caused comet tails to shorten. The decrease in comet tail abundance caused a proportional increase in actin clouds whereas overexpression of SM22 reversed the actin cloud to comet tail proportions and increased comet tail length, while not influencing EPEC pedestal abundance. Thus, we demonstrate that SM22 plays a role in regulating the transitions and morphological appearance of bacterially generated actin-rich structures during infections.


Assuntos
Actinas/metabolismo , Infecções Bacterianas/metabolismo , Estruturas Celulares/metabolismo , Estruturas Celulares/microbiologia , Proteínas dos Microfilamentos/fisiologia , Proteínas Musculares/fisiologia , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/patologia , Células CACO-2 , Células Cultivadas , Estruturas Celulares/patologia , Escherichia coli Enteropatogênica , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/metabolismo , Células HeLa , Humanos , Listeria monocytogenes , Listeriose/genética , Listeriose/metabolismo , Potoroidae
6.
PLoS One ; 9(8): e104881, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25115488

RESUMO

The highly infectious bacteria, Francisella tularensis, colonize a variety of organs and replicate within both phagocytic as well as non-phagocytic cells, to cause the disease tularemia. These microbes contain a conserved cluster of important virulence genes referred to as the Francisella Pathogenicity Island (FPI). Two of the most characterized FPI genes, iglC and pdpA, play a central role in bacterial survival and proliferation within phagocytes, but do not influence bacterial internalization. Yet, their involvement in non-phagocytic epithelial cell infections remains unexplored. To examine the functions of IglC and PdpA on bacterial invasion and replication during epithelial cell infections, we infected liver and lung epithelial cells with F. novicida and F. tularensis 'Type B' Live Vaccine Strain (LVS) deletion mutants (ΔiglC and ΔpdpA) as well as their respective gene complements. We found that deletion of either gene significantly reduced their ability to invade and replicate in epithelial cells. Gene complementation of iglC and pdpA partially rescued bacterial invasion and intracellular growth. Additionally, substantial LAMP1-association with both deletion mutants was observed up to 12 h suggesting that the absence of IglC and PdpA caused deficiencies in their ability to dissociate from LAMP1-positive Francisella Containing Vacuoles (FCVs). This work provides the first evidence that IglC and PdpA are important pathogenic factors for invasion and intracellular growth of Francisella in epithelial cells, and further highlights the discrete mechanisms involved in Francisella infections between phagocytic and non-phagocytic cells.


Assuntos
Francisella tularensis/genética , Francisella tularensis/patogenicidade , Francisella/genética , Francisella/patogenicidade , Ilhas Genômicas , Virulência/genética , Animais , Linhagem Celular , Células Epiteliais/microbiologia , Francisella/crescimento & desenvolvimento , Francisella tularensis/crescimento & desenvolvimento , Genes Bacterianos , Hepatócitos/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Pulmão/citologia , Pulmão/microbiologia , Proteínas de Membrana Lisossomal/metabolismo , Camundongos , Vacúolos/metabolismo , Vacúolos/microbiologia
7.
J Microbiol Methods ; 93(2): 153-60, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23523968

RESUMO

Francisella tularensis (F. tularensis), the causative agent of tularemia, has long been known to invade and occupy non-phagocytic epithelial cells. Many epithelial cell infection models have been developed to study this process; however, due to the lack of consensus on infection methods and precise experimental procedures to evaluate invasion and replication, selection of appropriate models to use based on the literature is challenging. To evaluate in vitro non-phagocytic cell infection models, we chose 8 epithelial cultured cell lines from published models to infect with F. tularensis subspecies novicida (F. novicida) and compared the results to a recently developed model that used the mouse hepatocyte BNL CL.2 cell line. We utilized classical gentamicin-based invasion assays to determine total intracellular bacterial loads and employed microscopic examination with staining techniques that distinguished between intracellular and extracellular bacteria to provide an accurate assessment of the proportion of invaded host cells and the degree of bacterial replication. We found that COS-7 cells exhibited the greatest invasion rates; CMT-93 cells contained the largest intracellular bacterial loads; ad HEK-293s were capable of invasion and replication rates at high levels, but required shorter infection incubation times. Although COS-7, CMT-93 and HEK-293 cell lines may be suited to study certain aspects of invasion or replication, we found that BNL CL.2 cells appeared the most appropriate to study the overall pathogenesis of F. novicida when examined in toto.


Assuntos
Endocitose , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Francisella tularensis/patogenicidade , Animais , Carga Bacteriana , Linhagem Celular , Citoplasma/microbiologia , Humanos
8.
Cell Microbiol ; 14(7): 1097-108, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22381134

RESUMO

The bacterial pathogens Listeria monocytogenes and enteropathogenic Escherichia coli (EPEC) generate motile actin-rich structures (comet tails and pedestals) as part of their infectious processes. Nexilin, an actin-associated protein and a component of focal adhesions, has been suggested to be involved in actin-based motility. To determine whether nexilin is commandeered during L. monocytogenes and EPEC infections, we infected cultured cells and found that nexilin is crucial for L. monocytogenes invasion as levels of internalized bacteria were significantly decreased in nexilin-targeted siRNA-treated cells. In addition, nexilin is a component of the machinery that drives the formation of L. monocytogenes comet tails and EPEC pedestals. Nexilin colocalizes with stationary bacteria and accumulates at the distal portion of comet tails and pedestals of motile bacteria. We also show that nexilin is crucial for efficient comet tail formation as cells pre-treated with nexilin siRNA generate malformed comet tails, whereas nexilin is dispensable during EPEC pedestal generation. These findings demonstrate that nexilin is required for efficient infection with invasive and adherent bacteria and is key to the actin-rich structures these microbes generate.


Assuntos
Actinas/metabolismo , Escherichia coli Enteropatogênica/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidade , Proteínas dos Microfilamentos/metabolismo , Linhagem Celular , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos
9.
Anat Rec (Hoboken) ; 295(2): 201-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22190417

RESUMO

Enteropathogenic Escherichia coli (EPEC) manipulate the cytoskeleton of host intestinal epithelial cells, producing membrane protrusions termed pedestals that the bacteria reside on throughout the course of their infections. By definition pedestals are actin-based structures, however recent work has identified the spectrin cytoskeleton as a necessary component of EPEC pedestals. Here, we investigated the detailed arrangement of the spectrin and actin cytoskeletons within these structures. Immunofluorescent imaging revealed that the spectrin network forms a peripheral cage around actin at the membranous regions of pedestals. Myosin S1 fragment decorated actin filaments examined by electron microscopy demonstrated that actin filaments orientate with their fast-growing barbed ends toward the lateral membranes of EPEC pedestals. These findings provide a detailed descriptive analysis, which further illustrate the spectrin cytoskeletal organization within these structures.


Assuntos
Extensões da Superfície Celular/ultraestrutura , Citoesqueleto/ultraestrutura , Escherichia coli Enteropatogênica/ultraestrutura , Células HeLa/ultraestrutura , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Animais , Extensões da Superfície Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Escherichia coli Enteropatogênica/metabolismo , Feminino , Técnica Indireta de Fluorescência para Anticorpo/métodos , Células HeLa/metabolismo , Células HeLa/microbiologia , Humanos , Masculino , Microscopia Eletrônica de Transmissão , Subfragmentos de Miosina/metabolismo , Ratos , Ratos Sprague-Dawley , Espectrina/metabolismo
10.
Microb Pathog ; 52(3): 149-56, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22197999

RESUMO

Recent work has demonstrated that the spectrin cytoskeleton is a host cell target, exploited during intestinal bacterial disease. Here we show that the highly virulent intestinal pathogen enterohaemorrhagic Escherichia coli (EHEC) is also reliant upon the spectrin cytoskeleton during key pathogenic events. Immunofluorescent microscopy demonstrated that the core components of the spectrin cytoskeleton (spectrin, adducin, and protein 4.1 [p4.1]) are recruited to sites of EHEC attachment and localized at pedestal structures along with the EHEC pedestal specific proteins IRSp53 and IRTKS. Further studies involving siRNA-mediated knockdowns of spectrin, adducin, or p4.1 revealed that those proteins are needed for efficient docking of EHEC to host cells, are involved in recruiting IRSp53 to the pedestal and are necessary for pedestal formation. These findings identify the spectrin cytoskeleton as a major host cell cytoskeletal network involved in critical EHEC pathogenic events.


Assuntos
Aderência Bacteriana , Proteínas de Ligação a Calmodulina/metabolismo , Escherichia coli Êntero-Hemorrágica/patogenicidade , Interações Hospedeiro-Patógeno , Proteínas de Membrana/metabolismo , Espectrina/metabolismo , Proteínas de Ligação a Calmodulina/antagonistas & inibidores , Inativação Gênica , Células HeLa , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espectrina/antagonistas & inibidores
11.
PLoS One ; 6(5): e19940, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21603579

RESUMO

Various enteric bacterial pathogens target the host cell cytoskeletal machinery as a crucial event in their pathogenesis. Despite thorough studies detailing strategies microbes use to exploit these components of the host cell, the role of the spectrin-based cytoskeleton has been largely overlooked. Here we show that the spectrin cytoskeleton is a host system that is hijacked by adherent (Entropathogenic Escherichia coli [EPEC]), invasive triggering (Salmonella enterica serovar Typhimurium [S. Typhimurium]) and invasive zippering (Listeria monocytogenes) bacteria. We demonstrate that spectrin cytoskeletal proteins are recruited to EPEC pedestals, S. Typhimurium membrane ruffles and Salmonella containing vacuoles (SCVs), as well as sites of invasion and comet tail initiation by L. monocytogenes. Spectrin was often seen co-localizing with actin filaments at the cell periphery, however a disconnect between the actin and spectrin cytoskeletons was also observed. During infections with S. Typhimurium ΔsipA, actin-rich membrane ruffles at characteristic sites of bacterial invasion often occurred in the absence of spectrin cytoskeletal proteins. Additionally, early in the formation of L. monocytogenes comet tails, spectrin cytoskeletal elements were recruited to the surface of the internalized bacteria independent of actin filaments. Further studies revealed the presence of the spectrin cytoskeleton during SCV and Listeria comet tail formation, highlighting novel cytoplasmic roles for the spectrin cytoskeleton. SiRNA targeted against spectrin and the spectrin-associated proteins severely diminished EPEC pedestal formation as well as S. Typhimurium and L. monocytogenes invasion. Ultimately, these findings identify the spectrin cytoskeleton as a ubiquitous target of enteric bacterial pathogens and indicate that this cytoskeletal system is critical for these infections to progress.


Assuntos
Aderência Bacteriana , Proteínas do Citoesqueleto , Infecções por Enterobacteriaceae/patologia , Enterobacteriaceae/patogenicidade , Interações Hospedeiro-Patógeno , Espectrina , Infecções por Enterobacteriaceae/etiologia , Escherichia coli/patogenicidade , Células HeLa , Humanos , Listeria monocytogenes/patogenicidade , Salmonella typhimurium/patogenicidade
12.
Sci Rep ; 1: 192, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22355707

RESUMO

Francisella tularensis are highly infectious microbes that cause the disease tularemia. Although much of the bacterial burden is carried in non-phagocytic cells, the strategies these pathogens use to invade these cells remains elusive. To examine these mechanisms we developed two in vitro Francisella-based infection models that recapitulate the non-phagocytic cell infections seen in livers of infected mice. Using these models we found that Francisella novicida exploit clathrin and cholesterol dependent mechanisms to gain entry into hepatocytes. We also found that the clathrin accessory proteins AP-2 and Eps15 co-localized with invading Francisella novicida as well as the Francisella Live Vaccine Strain (LVS) during hepatocyte infections. Interestingly, caveolin, a protein involved in the invasion of Francisella in phagocytic cells, was not required for non-phagocytic cell infections. These results demonstrate a novel endocytic mechanism adopted by Francisella and highlight the divergence in strategies these pathogens utilize between non-phagocytic and phagocytic cell invasion.


Assuntos
Colesterol/metabolismo , Clatrina/metabolismo , Endocitose/fisiologia , Francisella tularensis/fisiologia , Hepatócitos/microbiologia , Tularemia/microbiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Caveolinas/metabolismo , Linhagem Celular , Feminino , Hepatócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Estatísticos , Fagocitose , Pinocitose , Estrutura Terciária de Proteína , Interferência de RNA , Fator de Transcrição AP-2/metabolismo , Tularemia/patologia
13.
Protoplasma ; 244(1-4): 75-90, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20574860

RESUMO

Understanding the mechanisms that microbes exploit to invade host cells and cause disease is crucial if we are to eliminate their threat. Although pathogens use a variety of microbial factors to trigger entry into non-phagocytic cells, their targeting of the host cell process of endocytosis has emerged as a common theme. To accomplish this, microbes often rewire the normal course of particle internalization, frequently usurping theoretical maximal sizes to permit entry and reconfiguring molecular components that were once thought to be required for vesicle formation. Here, we discuss recent advances in our understanding of how toxins, viruses, bacteria, and fungi manipulate the host cell endocytic machinery to generate diseases. Additionally, we will reveal the advantages of using these organisms to expand our general knowledge of endocytic mechanisms in eukaryotic cells.


Assuntos
Bactérias/metabolismo , Endocitose , Fungos/metabolismo , Vírus/metabolismo , Animais , Humanos , Toxinas Biológicas/metabolismo
14.
Gut ; 59(2): 218-26, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19828465

RESUMO

OBJECTIVE: The attaching and effacing (A/E) pathogens enterohaemorrhagic Escherichia coli, enteropathogenic E coli and Citrobacter rodentium colonise intestinal tracts, attach to enterocytes, collapse infected cell microvilli and alter numerous host cell processes during infection. Enterocyte alterations result in numerous small molecules being released from host cells that likely contribute to diarrhoeal phenotypes observed during these infections. One possible route for small molecules to be released from intestinal cells may be through functional gap junction hemichannels. Here we examine the involvement of these hemichannels during the diarrhoeal disease caused by A/E pathogens in vivo. DESIGN: Mice were infected with the diarrhoea-causing murine A/E pathogen C rodentium for 7 days. Connexin43 (Cx43) protein levels and immunolocalization in the colon were initially used to determine alterations during A/E bacterial infections in vivo. Connexin mimetic peptides and connexin permeable tracer molecules were used to gage the presence and function of unpaired connexin hemichannels. The role of Cx43 in diarrhoea generation was assessed by comparing infections of wild-type mice to Cx43 mutant mice and determining the water abundance in the colonic luminal material. RESULTS: We demonstrate that Cx43 protein levels are increased in colonocytes during in vivo A/E bacterial infections, resulting in functionally open connexon hemichannels in apical membranes of infected cells. moreover, infected Cx43 +/- mice do not suffer from diarrhoeal disease. CONCLUSIONS: This study provides the first evidence that functional connexon hemichannels can occur in the intestine and are a novel molecular mechanism of water release during infectious diarrhoea.


Assuntos
Diarreia/microbiologia , Infecções por Enterobacteriaceae/complicações , Junções Comunicantes/fisiologia , Animais , Água Corporal/metabolismo , Citrobacter rodentium , Colo/metabolismo , Colo/ultraestrutura , Conexina 43/deficiência , Conexina 43/metabolismo , Diarreia/metabolismo , Diarreia/fisiopatologia , Infecções por Enterobacteriaceae/metabolismo , Corantes Fluorescentes , Genótipo , Isoquinolinas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Imunoeletrônica , Reação em Cadeia da Polimerase/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...