Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Microbiol Spectr ; 12(4): e0200623, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38358281

RESUMEN

The gram-positive bacterium Staphylococcus aureus can invade non-professional phagocytic cells by associating with the plasma protein fibronectin to exploit host cell integrins. Integrin-mediated internalization of these pathogens is facilitated by the local production of phosphatidylinositol-4,5-bisphosphate (PI-4,5-P2) via an integrin-associated isoform of phosphatidylinositol-5' kinase. In this study, we addressed the role of PI-4,5-P2-directed phosphatases on internalization of S. aureus. ShRNA-mediated knockdown of individual phosphoinositide 5-phosphatases revealed that synaptojanin1 (SYNJ1) is counteracting invasion of S. aureus into mammalian cells. Indeed, shRNA-mediated depletion as well as genetic deletion of synaptojanin1 via CRISPR/Cas9 resulted in a gain-of-function phenotype with regard to integrin-mediated uptake. Surprisingly, the surface level of integrins was slightly downregulated in Synj1-KO cells. Nevertheless, these cells showed enhanced local accumulation of PI-4,5-P2 and exhibited increased internalization of S. aureus. While the phosphorylation level of the integrin-associated protein tyrosine kinase FAK was unaltered, the integrin-binding and -activating protein talin was enriched in the vicinity of S. aureus in synaptojanin1 knockout cells. Scanning electron microscopy revealed enlarged membrane invaginations in the absence of synaptojanin1 explaining the increased capability of these cells to internalize integrin-bound microorganisms. Importantly, the enhanced uptake by Synj1-KO cells and the exaggerated morphological features were rescued by the re-expression of the wild-type enzyme but not phosphatase inactive mutants. Accordingly, synaptojanin1 activity limits integrin-mediated invasion of S. aureus, corroborating the important role of PI-4,5-P2 during this process.IMPORTANCEStaphylococcus aureus, an important bacterial pathogen, can invade non-professional phagocytes by capturing host fibronectin and engaging integrin α5ß1. Understanding how S. aureus exploits this cell adhesion receptor for efficient cell entry can also shed light on the physiological regulation of integrins by endocytosis. Previous studies have found that a specific membrane lipid, phosphatidylinositol-4,5-bisphosphate (PIP2), supports the internalization process. Here, we extend these findings and report that the local levels of PIP2 are controlled by the activity of the PIP2-directed lipid phosphatase Synaptojanin1. By dephosphorylating PIP2 at bacteria-host cell attachment sites, Synaptojanin1 counteracts the integrin-mediated uptake of the microorganisms. Therefore, our study not only generates new insight into subversion of cellular receptors by pathogenic bacteria but also highlights the role of host cell proteins acting as restriction factors for bacterial invasion at the plasma membrane.


Asunto(s)
Proteínas del Tejido Nervioso , Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Staphylococcus aureus/metabolismo , Integrinas/metabolismo , Fibronectinas/metabolismo , Fosfatidilinositoles/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , ARN Interferente Pequeño , Mamíferos
2.
Cell Host Microbe ; 27(5): 793-808.e5, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32289262

RESUMEN

Several pathogens suppress exfoliation, a key defense of epithelia against microbial colonization. Common among these pathogens, exemplified by Neisseria gonorrhoeae, is their ability to bind carcinoembryonic antigen-related cell adhesion molecules (CEACAMs). Gonococcal CEACAM engagement triggers the expression of CD105, which is necessary to block epithelial exfoliation, whereas homotypic CEACAM-CEACAM interactions or antibody-mediated CEACAM clustering does not lead to CD105 expression. Here, we show that CEACAM-associated bacteria release nitric oxide (NO) during anaerobic respiration, and membrane-permeable NO initiates a eukaryotic signaling pathway involving soluble guanylate cyclase (sGC), protein kinase G, and the transcription factor CREB to upregulate CD105 expression. A murine vaginal infection model with N. gonorrhoeae reveals this metabolic cross communication allows bacterial suppression of epithelial exfoliation to facilitate mucosal colonization. Disrupting NO-initiated responses in host cells re-establishes epithelial exfoliation and inhibits mouse genital tract colonization by N. gonorrhoeae, suggesting a host-directed approach to prevent bacterial infections.


Asunto(s)
Células Epiteliales/metabolismo , Gonorrea/metabolismo , Neisseria gonorrhoeae/metabolismo , Óxido Nítrico/metabolismo , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Antígenos CD , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Sitios de Unión , Antígeno Carcinoembrionario/metabolismo , Proteínas Portadoras , Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/microbiología , Epitelio , Femenino , Proteínas Ligadas a GPI , Gonorrea/microbiología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Neisseria gonorrhoeae/genética , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal
3.
PLoS Pathog ; 12(5): e1005608, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27171273

RESUMEN

Attachment to the host mucosa is a key step in bacterial pathogenesis. On the apical surface of epithelial cells, members of the human carcinoembryonic antigen (CEA) family are abundant glycoproteins involved in cell-cell adhesion and modulation of cell signaling. Interestingly, several gram-negative bacterial pathogens target these receptors by specialized adhesins. The prototype of a CEACAM-binding pathogen, Neisseria gonorrhoeae, utilizes colony opacity associated (Opa) proteins to engage CEA, as well as the CEA-related cell adhesion molecules CEACAM1 and CEACAM6 on human epithelial cells. By heterologous expression of neisserial Opa proteins in non-pathogenic E. coli we find that the Opa protein-CEA interaction is sufficient to alter gene expression, to increase integrin activity and to promote matrix adhesion of infected cervical carcinoma cells and immortalized vaginal epithelial cells in vitro. These CEA-triggered events translate in suppression of exfoliation and improved colonization of the urogenital tract by Opa protein-expressing E. coli in CEA-transgenic compared to wildtype mice. Interestingly, uropathogenic E. coli expressing an unrelated CEACAM-binding protein of the Afa/Dr adhesin family recapitulate the in vitro and in vivo phenotype. In contrast, an isogenic strain lacking the CEACAM-binding adhesin shows reduced colonization and does not suppress epithelial exfoliation. These results demonstrate that engagement of human CEACAMs by distinct bacterial adhesins is sufficient to blunt exfoliation and to promote host infection. Our findings provide novel insight into mucosal colonization by a common UPEC pathotype and help to explain why human CEACAMs are a preferred epithelial target structure for diverse gram-negative bacteria to establish a foothold on the human mucosa.


Asunto(s)
Adhesión Bacteriana/fisiología , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Membrana Mucosa/microbiología , Sistema Urogenital/microbiología , Escherichia coli Uropatógena/patogenicidad , Animales , Western Blotting , Línea Celular Tumoral , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Membrana Mucosa/metabolismo , Sistema Urogenital/metabolismo
4.
Infect Immun ; 81(7): 2358-70, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23630956

RESUMEN

Members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family serve as cellular receptors for Neisseria gonorrhoeae. More specifically, neisserial colony opacity (OpaCEA)) proteins bind to epithelial CEACAMs (CEACAM1, CEA, CEACAM6) to promote bacterial colonization of the mucosa. In contrast, recognition by CEACAM3, expressed by human granulocytes, results in uptake and destruction of Opa(CEA)-expressing bacteria. Therefore, CEACAM3-mediated uptake might limit the spread of gonococci. However, some strains can cause disseminating gonococcal infections (DGIs), and it is currently unknown how these strains escape detection by granulocyte CEACAM3. Therefore, the opa gene loci from N. gonorrhoeae strain VP1, which was derived from a patient with disseminated gonococcal disease, were cloned and constitutively expressed in Escherichia coli. Similar to Opa proteins of the nondisseminating strain MS11, the majority of Opa proteins from strain VP1 bound epithelial CEACAMs and promoted CEACAM-initiated responses by epithelial cells. In sharp contrast to the Opa proteins of strain MS11, the Opa proteins of strain VP1 failed to interact with the human granulocyte receptor CEACAM3. Accordingly, bacteria expressing VP1 Opa proteins were not taken up by primary human granulocytes and did not trigger a strong oxidative burst. Analysis of Opa variants from four additional clinical DGI isolates again demonstrated a lack of CEACAM3 binding. In summary, our results reveal that particular N. gonorrhoeae strains express an Opa protein repertoire allowing engagement of epithelial CEACAMs for successful mucosal colonization, while avoiding recognition and elimination via CEACAM3-mediated phagocytosis. A failure of CEACAM3-mediated innate immune detection might be linked to the ability of gonococci to cause disseminated infections.


Asunto(s)
Granulocitos/inmunología , Inmunidad Innata , Neisseria gonorrhoeae/patogenicidad , Neutrófilos/inmunología , Alelos , Secuencia de Aminoácidos , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Clonación Molecular , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Escherichia coli/genética , Escherichia coli/metabolismo , Sitios Genéticos , Gonorrea/inmunología , Gonorrea/microbiología , Granulocitos/microbiología , Células HEK293 , Humanos , Datos de Secuencia Molecular , Neisseria gonorrhoeae/inmunología , Neutrófilos/microbiología , Fagocitosis , Unión Proteica , Especificidad de la Especie
5.
Curr Opin Cell Biol ; 24(5): 637-44, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22884865

RESUMEN

Numerous pathogens express adhesive proteins to directly or indirectly associate with integrins. It is well established that by targeting integrins, microbes not only establish an intimate contact with host tissues, but also trigger cellular responses including bacterial internalization. This review will summarize current knowledge about the role of these integrin-dependent processes during infection and how bacteria assure that they efficiently connect to integrins for host cell invasion or translocation of effector molecules. Furthermore, we will discuss recent insight demonstrating that bacteria can harness the physiological, matrix-binding function of integrins for promoting host colonization. From these combined studies, it is becoming evident that integrins are a common nexus for the manipulation of cellular functions by bacterial pathogens. Approaches to disrupt this connection might be an appropriate means to obtain broad-acting tools to modulate a spectrum of infectious diseases.


Asunto(s)
Bacterias/patogenicidad , Integrinas/metabolismo , Adhesinas Bacterianas/metabolismo , Animales , Bacterias/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Humanos , Ligandos
6.
Science ; 329(5996): 1197-201, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20813953

RESUMEN

Colonization of mucosal surfaces is the key initial step in most bacterial infections. One mechanism protecting the mucosa is the rapid shedding of epithelial cells, also termed exfoliation, but it is unclear how pathogens counteract this process. We found that carcinoembryonic antigen (CEA)-binding bacteria colonized the urogenital tract of CEA transgenic mice, but not of wild-type mice, by suppressing exfoliation of mucosal cells. CEA binding triggered de novo expression of the transforming growth factor receptor CD105, changing focal adhesion composition and activating beta1 integrins. This manipulation of integrin inside-out signaling promotes efficient mucosal colonization and represents a potential target to prevent or cure bacterial infections.


Asunto(s)
Antígeno Carcinoembrionario/metabolismo , Células Epiteliales/patología , Gonorrea/microbiología , Cadenas beta de Integrinas/metabolismo , Neisseria gonorrhoeae/metabolismo , Neisseria gonorrhoeae/patogenicidad , Vagina/microbiología , Animales , Antígenos Bacterianos/metabolismo , Antígenos CD/metabolismo , Antígeno Carcinoembrionario/genética , Proteínas del Citoesqueleto/metabolismo , Endoglina , Células Epiteliales/microbiología , Femenino , Adhesiones Focales , Proteínas Ligadas a GPI , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Membrana Mucosa/microbiología , Neisseria gonorrhoeae/aislamiento & purificación , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transfección , Vagina/citología , Vagina/patología , Zixina
7.
Cell Microbiol ; 10(5): 1074-92, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18081725

RESUMEN

Several bacterial pathogens exploit carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) to promote attachment and uptake into eukaryotic host cells. The widely expressed isoform CEACAM1 is involved in cell-cell adhesion, regulation of cell proliferation, insulin homeostasis, and neo-angiogenesis, processes that depend on the cytoplasmic domain of CEACAM1. By analysing the molecular requirements for CEACAM1-mediated internalization of bacteria, we surprisingly find that the CEACAM1 cytoplasmic domain is completely obsolete for bacterial uptake. Accordingly, CEACAM1-4L as well as a CEACAM1 mutant with a complete deletion of the cytoplasmic domain (CEACAM1 DeltaCT) promote equivalent internalization of several human pathogens. CEACAM1-4L- and CEACAM1 DeltaCT-mediated uptake proceeds in the presence of inhibitors of actin microfilament dynamics, which is in contrast to CEACAM3-mediated internalization. Bacteria-engaged CEACAM1-4L and CEACAM1 DeltaCT, but not CEACAM3, localize to a gangliosid GM1- and GPI-anchored protein-containing portion of the plasma membrane. In addition, interference with cholesterol-rich membrane microdomains severely blocks bacterial uptake via CEACAM1-4L and CEACAM1 DeltaCT, but not CEACAM3. Similar to GPI-anchored CEACAM6, both CEACAM1-4L as well as CEACAM1 DeltaCT partition into a low-density, Triton-insoluble membrane fraction upon receptor clustering, whereas CEACAM3 is not detected in this fraction. Bacterial uptake by truncated CEACAM1 or chimeric CEACAM1/CEACAM3 molecules reveals that the transmembrane domain of CEACAM1 is responsible for its association with membrane microdomains. Together, these data argue for a functional role of lipid rafts in CEACAM1-mediated endocytosis that is promoted by the transmembrane domain of the receptor and that might be relevant for CEACAM1 function in physiologic settings.


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Microdominios de Membrana/metabolismo , Neisseria gonorrhoeae/fisiología , Actinas/metabolismo , Antígenos CD/química , Moléculas de Adhesión Celular/química , Línea Celular , Colesterol/metabolismo , Citometría de Flujo , Humanos , Microscopía Electrónica de Rastreo , Fosforilación , Estructura Terciaria de Proteína , Tirosina/metabolismo
8.
Eur J Cell Biol ; 85(3-4): 235-42, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16546567

RESUMEN

A large number of bacterial pathogens targets cell adhesion molecules to establish an intimate contact with host cells and tissues. Members of the integrin, cadherin and immunoglobulin-related cell adhesion molecule (IgCAM) families are frequently recognized by specific bacterial surface proteins. Binding can trigger bacterial internalization following cytoskeletal rearrangements that are initiated upon receptor clustering. Moreover, signals emanating from the occupied receptors can result in cellular responses such as gene expression events that influence the phenotype of the infected cell. This review will address recent advances in our understanding of bacterial engagement of cellular adhesion molecules by discussing the binding of integrins by Staphylococcus aureus as well as the exploitation of IgCAMs by pathogenic Neisseria species.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Infecciones por Bacterias Gramnegativas/microbiología , Neisseria/patogenicidad , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Humanos , Inmunoglobulinas/inmunología , Inmunoglobulinas/metabolismo , Integrinas/metabolismo , Neisseria/citología , Neisseria/metabolismo , Staphylococcus aureus/citología , Staphylococcus aureus/metabolismo
9.
J Cell Biol ; 170(5): 825-36, 2005 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-16115956

RESUMEN

Exfoliation, which is the detachment of infected epithelial cells, is an innate defense mechanism to prevent bacterial colonization. Indeed, infection with Neisseria gonorrhoeae induced epithelial detachment from an extracellular matrix (ECM) substrate in vitro. Surprisingly, variants of N. gonorrhoeae that bind to human carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) failed to induce detachment and, instead, promoted enhanced host cell adhesion to the ECM. Microarray analysis revealed that CEACAM engagement by several human pathogens triggers expression of CD105. Blockage of CD105 expression by antisense oligonucleotides abolished infection-induced cell adhesion. The expression of full-length CD105 promoted cell adhesion to the ECM and was sufficient to prevent infection-induced detachment. The CD105-mediated increase in cell adhesion was dependent on the presence and function of integrin beta1. CD105 expression did not elevate cellular integrin levels but caused a dramatic increase in the ECM-binding capacity of the cells, suggesting that CD105 affects integrin activity. The exploitation of CEACAMs to trigger CD105 expression and to counteract infection-induced cell detachment represents an intriguing adaptation of pathogens that are specialized to colonize the human mucosa.


Asunto(s)
Antígenos CD/metabolismo , Adhesión Celular/fisiología , Células Epiteliales/metabolismo , Neisseria gonorrhoeae/patogenicidad , Animales , Antígenos Bacterianos/metabolismo , Antígenos CD/genética , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Endoglina , Células Epiteliales/citología , Matriz Extracelular/metabolismo , Humanos , Cadenas alfa de Integrinas/metabolismo , Integrina beta1/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Superficie Celular , Transducción de Señal/fisiología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
Biol Chem ; 386(5): 481-90, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15927892

RESUMEN

Neisseria gonorrhoeae (Ngo) is a Gram-negative pathogenic bacterium responsible for an array of diseases ranging from urethritis to disseminated gonococcal infections. Early events in the establishment of infection involve interactions between Ngo and the mucosal epithelium, which induce a local inflammatory response. Here we analyzed the molecular mechanism involved in the Ngo-induced induction of the proinflammatory cytokines tumor necrosis factor alpha (TNFalpha), interleukin-6 (IL-6), and IL-8. We identified the immediate early response transcription factor nuclear factor kappaB (NF-kappaB) as a key molecule for the induction of cytokine release. Ngo-induced activation of direct upstream signaling molecules was demonstrated for IkappaB kinase alpha and beta (IKKalpha and IKKbeta) by phosphorylation of IkappaBalpha as a substrate and IKK autophosphorylation. Using dominant negative cDNAs encoding kinase-dead IKKalpha, IKKbeta, and NF-kappaB-inducing kinase (NIK), Ngo-induced NF-kappaB activity was significantly inhibited. Curcumin, the yellow pigment derived from Curcuma longa, inhibited IKKalpha, IKKbeta and NIK, indicating its strong potential to block NF-kappaB-mediated cytokine release and the innate immune response. In addition to the inhibition of Ngo-induced signaling, curcumin treatment of cells completely abolished the adherence of bacteria to cells in late infection, underlining the high potential of curcumin as an anti-microbial compound without cytotoxic side effects.


Asunto(s)
Antiinflamatorios/farmacología , Adhesión Bacteriana/efectos de los fármacos , Curcumina/farmacología , Citocinas/antagonistas & inhibidores , Gonorrea/metabolismo , FN-kappa B/antagonistas & inhibidores , Neisseria gonorrhoeae/efectos de los fármacos , Quimiocinas/antagonistas & inhibidores , Quimiocinas/metabolismo , Citocinas/metabolismo , Activación Enzimática , Gonorrea/microbiología , Células HeLa , Humanos , FN-kappa B/química , FN-kappa B/fisiología , Neisseria gonorrhoeae/fisiología , Transducción de Señal/efectos de los fármacos , Activación Transcripcional
11.
J Biol Chem ; 277(9): 7438-46, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11751883

RESUMEN

The human-specific pathogen Neisseria gonorrhoeae expresses opacity-associated (Opa) protein adhesins that bind to various members of the carcinoembryonic antigen-related cellular adhesion molecule (CEACAM) family. In this study, we have analyzed the mechanism underlying N. gonorrhoeae-induced CEACAM up-regulation in epithelial cells. Epithelial cells represent the first barrier for the microbial pathogen. We therefore characterized CEACAM expression in primary human ovarian surface epithelial (HOSE) cells and found that CEACAM1-3 (L, S) and CEACAM1-4 (L, S) splice variants mediate an increased Opa(52)-dependent gonoccocal binding to HOSE cells. Up-regulation of these CEACAM molecules in HOSE cells is a direct process that takes place within 2 h postinfection and depends on close contact between microbial pathogen and HOSE cells. N. gonorrhoeae-triggered CEACAM1 up-regulation involves activation of the transcription factor nuclear factor kappaB (NF-kappaB), which translocates as a p50/p65 heterodimer into the nucleus, and an NF-kappaB-specific inhibitory peptide inhibited CEACAM1-receptor up-regulation in N. gonorrhoeae-infected HOSE cells. Bacterial lipopolysaccharides did not induce NF-kappaB and CEACAM up-regulation, which corresponds to our findings that HOSE cells do not express toll-like receptor 4. The ability of N. gonorrhoeae to up-regulate its epithelial receptor CEACAM1 through NF-kappaB suggests an important mechanism allowing efficient bacterial colonization during the initial infection process.


Asunto(s)
Antígenos CD/biosíntesis , Antígenos de Diferenciación/biosíntesis , Células Epiteliales/metabolismo , FN-kappa B/metabolismo , Neisseria gonorrhoeae/metabolismo , Empalme Alternativo , Western Blotting , Moléculas de Adhesión Celular , Línea Celular , Núcleo Celular/metabolismo , Separación Celular , Citosol/metabolismo , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Microscopía Confocal , Ovario/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...