Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nat Commun ; 11(1): 483, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980623

RESUMO

Inflammatory bowel disease is associated with changes in the mucosal barrier, increased intestinal permeability, and increased risk of infections and sepsis, but the underlying mechanisms are incompletely understood. Here, we show how continuous translocation of gut microbial components affects iron homeostasis and facilitates susceptibility to inflammation-associated sepsis. A sub-lethal dose of lipopolysaccharide results in higher mortality in Mucin 2 deficient (Muc2-/-) mice, and is associated with elevated circulatory iron load and increased bacterial translocation. Translocation of gut microbial components attenuates hepatic stearoyl CoA desaturase-1 activity, a key enzyme in hepatic de novo lipogenesis. The resulting reduction of hepatic saturated and unsaturated fatty acid levels compromises plasma membrane fluidity of red blood cells, thereby significantly reducing their life span. Inflammation in Muc2-/- mice alters erythrophagocytosis efficiency of splenic macrophages, resulting in an iron-rich milieu that promotes bacterial growth. Our study thus shows that increased intestinal permeability triggers a cascade of events resulting in increased bacterial growth and risk of sepsis.


Assuntos
Mucosa Intestinal/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Sepse/metabolismo , Estearoil-CoA Dessaturase/metabolismo , Animais , Permeabilidade da Membrana Celular , Citofagocitose , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/microbiologia , Mucosa Intestinal/microbiologia , Ferro/sangue , Lipogênese , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mucina-2/deficiência , Mucina-2/genética , Sepse/etiologia , Sepse/microbiologia
2.
Nat Commun ; 10(1): 4306, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31541089

RESUMO

The mucus layer is the first line of innate host defense in the gut that protects the epithelium by spatially separating commensal bacteria. MUC2 mucin is produced and stored by goblet cells that is constitutively exocytosed or hyper secreted upon sensing a threat. How coordinated mucus exocytosis maintains homeostasis in the intestinal epithelium and modulates the immunological landscape remains elusive. Here we describe how the vesicle SNARE protein VAMP8 coordinates mucin exocytosis from goblet cells. Vamp8-/- exhibit a mild pro-inflammatory state basally due to an altered mucus layer and increased encounters with microbial antigens. Microbial diversity shifts to a detrimental microbiota with an increase abundance of pathogenic and mucolytic bacteria. To alleviate the heavy microbial burden and inflammatory state basally, Vamp8-/- skews towards tolerance. Despite this, Vamp8-/- is highly susceptible to both chemical and infectious colitis demonstrating the fragility of the intestinal mucosa without proper mucus exocytosis mechanisms.


Assuntos
Colo/metabolismo , Exocitose/fisiologia , Células Caliciformes/metabolismo , Mucosa Intestinal/metabolismo , Mucina-2/metabolismo , Proteínas R-SNARE/metabolismo , Animais , Biodiversidade , Colo/patologia , Citocinas/metabolismo , Células Caliciformes/patologia , Homeostase , Humanos , Intestinos/patologia , Camundongos Knockout , Microbiota , Mucina-2/genética , Muco/metabolismo , Fenótipo , Proteínas R-SNARE/genética , Proteínas SNARE/metabolismo
3.
Am J Pathol ; 188(6): 1354-1373, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29545196

RESUMO

MUC2 mucin is a large glycoprotein produced by goblet cells that forms the protective mucus blanket overlying the intestinal epithelium as the first line of innate host defense. High MUC2 production in inflammatory bowel disease and infectious colitis depletes goblet cells and the mucus layer by an unknown mechanism. Herein, we analyzed the effect of high MUC2 biosynthesis on endoplasmic reticulum (ER) stress and apoptosis in goblet cells using a high MUC2-producing human goblet cell line (HT29-H) and an HT29-H clone (HT29-L) silenced for MUC2 expression by lentivirus-mediated shRNA. Goblet cell ER stress and apoptosis were quantified during early onset of dextran sulfate sodium-induced colitis in C57BL/6 and Math1M1GFP mice. Compared with HT29-L and MUC2 nonproducing Caco-2 cells, high MUC2-producing HT29-H cells had significantly increased ER stress and apoptosis after treatment with ER stress-inducing agents. Apoptosis was driven by increased misfolded MUC2 that triggered elevated levels of reactive oxygen species. Correcting MUC2 folding and inhibiting reactive oxygen species alleviated ER stress and rescued cells from apoptosis. During early-onset colitis, mucus hypersecretion caused severe ER stress and apoptosis of goblet cells that preceded absorptive epithelial cell damage. Thus, in gastrointestinal inflammation, high MUC2 biosynthesis and goblet cell apoptosis lead to a dysfunctional epithelial barrier. Enhancing MUC2 folding may help alleviate goblet cell depletion and maintain mucosal integrity.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Células Caliciformes/patologia , Mucina-2/química , Mucina-2/metabolismo , Dobramento de Proteína , Espécies Reativas de Oxigênio/metabolismo , Animais , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Sulfato de Dextrana/toxicidade , Células Caliciformes/metabolismo , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mucina-2/genética
4.
Microb Cell ; 4(12): 426-427, 2017 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-29234672

RESUMO

The intestinal mucosa encounters a barrage of ingested insults within the host yet under homeostasis elegantly facilitates nutrient absorption and sustenance of the commensal microbiota. An essential defence mechanism employed by the host is limiting the spatial niche various microbes may occupy as executed by the fluid mucus layer. Pathogens that violate their restricted niche within the intestinal mucosa are first expelled by robust mucus secretion from goblet cells thus by-passing the need for an immune response. Surprisingly, while many pathogens are known to exert hyper-secretion of mucus from goblet cells, the mechanisms governing this event remain elusive. In a recent report by Cornick et al (MBio 8: e01323-17), we nominate SNARE-mediated exocytosis as the putative mechanism responsible for pathogen-induced mucus secretion from goblet cells. The vesicle SNARE VAMP8 on mucin granules within goblet cells is specifically activated following infection with the protozoan parasite Entamoeba histolytica that is known to induce potent hyper-secretion and coordinates mucin exocytosis. This secretion event is critical in fending off a pathogen, as cells lacking VAMP8 are prone to increased E. histolytica colonization and cytolysis through apoptosis. Failing coordinated mucus exocytosis and subsequent epithelial barrier destruction, the host mounts an immune response as a last line of defence.

5.
mBio ; 8(5)2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28974617

RESUMO

Intestinal mucus secretion is critical in maintaining mucosal host defense against a myriad of pathogens by preventing direct association with the epithelium. Entamoeba histolytica specifically binds colonic MUC2 mucin and also induces potent hypersecretion from goblet cells; however, characterization of the nature of the mechanisms controlling mucus release remains elusive. In this report, we identify vesicle SNARE vesicle-associated membrane protein 8 (VAMP8) present on mucin granules as orchestrating regulated exocytosis in human goblet cells in response to the presence of E. histolytica VAMP8 was specifically activated during E. histolytica infection, and ablation of VAMP8 led to impaired mucin secretion. As a consequence, loss of VAMP8 increased E. histolytica adherence to epithelial cells associated with enhanced cell death through apoptosis characterized by caspase 3 and 9 cleavages and DNA fragmentation. With the mucosal barrier compromised in Vamp8-/- animals, E. histolytica induced an aggressive proinflammatory response with elevated levels of interleukin-1 alpha (IL-1α), IL-1ß, and tumor necrosis factor alpha (TNF-α) secretion. This report is the first to characterize regulated mucin exocytosis in intestinal goblet cells in response to a pathogen and the downstream consequences of improper mucin secretion in mucosal barrier defense.IMPORTANCE The intestinal tract is exposed to countless substances and pathogens, and yet homeostasis is maintained, in part by the mucus layer that houses the microbiota and spatially separates potential threats from the underlying single layer of epithelium. Despite the critical role of mucus in innate host defense, characterization of the mechanisms by which mucus is secreted from specialized goblet cells in the gut remains elusive. Here, we describe the machinery that regulates mucus secretion as well as the consequence during infection with the colonic pathogen Entamoeba histolytica Abolishment of the key machinery protein VAMP8 abrogated mucus release in cultured human colonic goblet cells and during E. histolytica infection in Vamp8-/- mice, which showed enhanced amoeba contact and killing of epithelial cells, triggering a potent proinflammatory response. This report highlights the importance of the VAMP8 secretory machinery in facilitating mucus release from intestinal goblet cells and the dire consequences that occur during disease pathogenesis if these pathways are not functional.


Assuntos
Entamoeba histolytica/fisiologia , Exocitose , Células Caliciformes/fisiologia , Imunidade Inata , Mucinas/metabolismo , Proteínas R-SNARE/metabolismo , Animais , Linhagem Celular , Entamoeba histolytica/patogenicidade , Células Epiteliais , Células Caliciformes/microbiologia , Camundongos , Mucina-2/metabolismo , Proteínas R-SNARE/deficiência , Proteínas R-SNARE/genética
6.
PLoS Pathog ; 13(8): e1006592, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28837696

RESUMO

Entamoeba histolytica (Eh) is the causative agent of amebiasis, one of the major causes of dysentery-related morbidity worldwide. Recent studies have underlined the importance of the intercellular junction between Eh and host cells as a determinant in the pathogenesis of amebiasis. Despite the fact that direct contact and ligation between Eh surface Gal-lectin and EhCP-A5 with macrophage α5ß1 integrin are absolute requirements for NLRP3 inflammasome activation and IL-1ß release, many other undefined molecular events and downstream signaling occur at the interface of Eh and macrophage. In this study, we investigated the molecular events at the intercellular junction that lead to recognition of Eh through modulation of the macrophage cytoskeleton. Upon Eh contact with macrophages key cytoskeletal-associated proteins were rapidly post-translationally modified only with live Eh but not with soluble Eh proteins or fragments. Eh ligation with macrophages rapidly activated caspase-6 dependent cleavage of the cytoskeletal proteins talin, Pyk2 and paxillin and caused robust release of the pro-inflammatory cytokine, IL-1ß. Macrophage cytoskeletal cleavages were dependent on Eh cysteine proteinases EhCP-A1 and EhCP-A4 but not EhCP-A5 based on pharmacological blockade of Eh enzyme inhibitors and EhCP-A5 deficient parasites. These results unravel a model where the intercellular junction between macrophages and Eh form an area of highly interacting proteins that implicate the macrophage cytoskeleton as a sensor for Eh contact that leads downstream to subsequent inflammatory immune responses.


Assuntos
Citoesqueleto/imunologia , Entamebíase/imunologia , Interações Hospedeiro-Parasita/imunologia , Interleucina-1beta/metabolismo , Macrófagos/imunologia , Macrófagos/parasitologia , Animais , Western Blotting , Linhagem Celular , Entamoeba histolytica/imunologia , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Microscopia Confocal
7.
Tissue Barriers ; 5(1): e1283386, 2017 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-28452682

RESUMO

Entamoeba histolytica (Eh) is the protozoan parasite responsible for intestinal amebiasis and interacts dynamically with the host intestinal epithelium during disease pathogenesis. A multifaceted pathogenesis profile accounts for why 90% of individuals infected with Eh are largely asymptomatic. For 100 millions individuals that are infected each year, key interactions within the intestinal mucosa dictate disease susceptibility. The ability for Eh to induce amebic colitis and disseminate into extraintestinal organs depends on the parasite competing with indigenous bacteria and overcoming the mucus barrier, binding to host cells inducing their cell death, invasion through the mucosa and outsmarting the immune system. In this review we summarize how Eh interacts with the intestinal epithelium and subverts host defense mechanisms in disease pathogenesis.


Assuntos
Entamoeba histolytica/patogenicidade , Interações Hospedeiro-Parasita/imunologia , Colo/metabolismo , Humanos , Mucosa Intestinal/metabolismo
8.
PLoS Pathog ; 12(4): e1005579, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27073869

RESUMO

Critical to the pathogenesis of intestinal amebiasis, Entamoeba histolytica (Eh) induces mucus hypersecretion and degrades the colonic mucus layer at the site of invasion. The parasite component(s) responsible for hypersecretion are poorly defined, as are regulators of mucin secretion within the host. In this study, we have identified the key virulence factor in live Eh that elicits the fast release of mucin by goblets cells as cysteine protease 5 (EhCP5) whereas, modest mucus secretion occurred with secreted soluble EhCP5 and recombinant CP5. Coupling of EhCP5-αvß3 integrin on goblet cells facilitated outside-in signaling by activating SRC family kinases (SFK) and focal adhesion kinase that resulted in the activation/phosphorlyation of PI3K at the site of Eh contact and production of PIP3. PKCδ was activated at the EhCP5-αvß3 integrin contact site that specifically regulated mucin secretion though the trafficking vesicle marker myristoylated alanine-rich C-kinase substrate (MARCKS). This study has identified that EhCP5 coupling with goblet cell αvß3 receptors can initiate a signal cascade involving PI3K, PKCδ and MARCKS to drive mucin secretion from goblet cells critical in disease pathogenesis.


Assuntos
Cisteína Proteases/metabolismo , Disenteria Amebiana/metabolismo , Entamoeba histolytica/patogenicidade , Células Caliciformes/metabolismo , Integrina alfaVbeta3/metabolismo , Mucinas/metabolismo , Animais , Western Blotting , Linhagem Celular , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Modelos Animais de Doenças , Disenteria Amebiana/patologia , Ensaio de Imunoadsorção Enzimática , Exocitose , Citometria de Fluxo , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Microscopia Confocal , Proteínas de Protozoários/metabolismo , Transdução de Sinais/fisiologia , Virulência/fisiologia , Fatores de Virulência/metabolismo
9.
PLoS Pathog ; 11(5): e1004887, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25955828

RESUMO

Entamoeba histolytica (Eh) is an extracellular protozoan parasite of humans that invades the colon to cause life-threatening intestinal and extra-intestinal amebiasis. Colonized Eh is asymptomatic, however, when trophozoites adhere to host cells there is a considerable inflammatory response that is critical in the pathogenesis of amebiasis. The host and/or parasite factors that trigger the inflammatory response to invading Eh are not well understood. We recently identified that Eh adherence to macrophages induces inflammasome activation and in the present study we sought to determine the molecular events upon contact that coordinates this response. Here we report that Eh contact-dependent activation of α5ß1 integrin is critical for activation of the NLRP3 inflammasome. Eh-macrophage contact triggered recruitment of α5ß1 integrin and NLRP3 into the intercellular junction, where α5ß1 integrin underwent activation by an integrin-binding cysteine protease on the parasite surface, termed EhCP5. As a result of its activation, α5ß1 integrin induced ATP release into the extracellular space through opening of pannexin-1 channels that signalled through P2X7 receptors to deliver a critical co-stimulatory signal that activated the NLRP3 inflammasome. Both the cysteine protease activity and integrin-binding domain of EhCP5 were required to trigger α5ß1 integrin that led to ATP release and NLRP3 inflammasome activation. These findings reveal engagement of α5ß1 integrin across the parasite-host junction is a key regulatory step that initiates robust inflammatory responses to Eh. We propose that α5ß1 integrin distinguishes Eh direct contact and functions with NLRP3 as pathogenicity sensor for invasive Eh infection.


Assuntos
Proteínas de Transporte/metabolismo , Entamoeba histolytica/imunologia , Entamebíase/metabolismo , Interações Hospedeiro-Patógeno , Inflamassomos/metabolismo , Integrina alfa5beta1/agonistas , Macrófagos/metabolismo , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Adesão Celular , Células Cultivadas , Cisteína Proteases/química , Cisteína Proteases/genética , Cisteína Proteases/metabolismo , Entamoeba histolytica/fisiologia , Entamebíase/imunologia , Entamebíase/parasitologia , Humanos , Imunidade Inata , Inflamassomos/imunologia , Integrina alfa5beta1/antagonistas & inibidores , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/parasitologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Proteína 3 que Contém Domínio de Pirina da Família NLR , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Proteólise , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Interferência de RNA , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
10.
Tissue Barriers ; 3(1-2): e982426, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25838985

RESUMO

The gastrointestinal tract is coated by a thick layer of mucus that forms the front line of innate host defense. Mucus consists of high molecular weight glycoproteins called mucins that are synthesized and secreted by goblet cells and functions primarily to lubricate the epithelium and protect it from damage by noxious substances. Recent studies have also suggested the involvement of goblet cells and mucins in complex immune functions such as antigen presentation and tolerance. Under normal physiological conditions, goblet cells continually produce mucins to replenish and maintain the mucus barrier; however, goblet cell function can be disrupted by various factors that can affect the integrity of the mucus barrier. Some of these factors such as microbes, microbial toxins and cytokines can stimulate or inhibit mucin production and secretion, alter the chemical composition of mucins or degrade the mucus layer. This can lead to a compromised mucus barrier and subsequently to various pathological conditions like chronic inflammatory diseases. Insight into how these factors modulate the mucus barrier in the gut is necessary in order to develop strategies to combat these disorders.

11.
PLoS One ; 8(9): e73339, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058468

RESUMO

The exact mechanism by which Entamoeba histolytica disrupts the human colonic epithelium and invades the mucosa has yet to be clearly elucidated. E. histolytica produces a diverse array of putative virulent factors such as glycosidase, cysteine proteinases and amebapore that can modulate and/or disrupt epithelial barrier functions. However, it is currently thought that E. histolytica produces numerous other molecules and strategies to disrupt colonic mucosal defenses. In this study, we document a putative mechanism whereby the parasite alters the integrity of human epithelium by expressing a cognate tight junction protein of the host. We detected this protein as "occludin-like" as revealed by immunoblotting and immunoprecipitation studies and visualization by confocal microscopy using antibodies highly specific for human occludin. We propose that E. histolytica occludin-like protein might displace mucosal epithelial occludin-occludin tight junction interactions resulting in epithelial disruption analogous to sub mucosal human dendritic cells sampling luminal contents. These results indicate that E. histolytica occludin is a putative virulent component that can play a role in the pathogenesis of intestinal amebiasis.


Assuntos
Entamoeba histolytica/genética , Células Epiteliais/parasitologia , Ocludina/genética , Proteínas de Protozoários/genética , Junções Íntimas/parasitologia , Fatores de Virulência/genética , Western Blotting , Linhagem Celular , Colo/metabolismo , Colo/parasitologia , Colo/patologia , Entamoeba histolytica/metabolismo , Entamoeba histolytica/patogenicidade , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação da Expressão Gênica , Interações Hospedeiro-Parasita , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitologia , Mucosa Intestinal/patologia , Microscopia Confocal , Ocludina/metabolismo , Permeabilidade , Proteínas de Protozoários/metabolismo , Especificidade da Espécie , Junções Íntimas/metabolismo , Junções Íntimas/patologia , Fatores de Virulência/metabolismo
12.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA